1
|
Hunjan G, Shah SS, Kosey S, Aran KR. Gut microbiota and the tryptophan-kynurenine pathway in anxiety: new insights and treatment strategies. J Neural Transm (Vienna) 2025:10.1007/s00702-025-02938-8. [PMID: 40369368 DOI: 10.1007/s00702-025-02938-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Accepted: 04/28/2025] [Indexed: 05/16/2025]
Abstract
Anxiety disorders are mental health disorders characterized by long-lasting fear, worry, nervousness, and alterations in gut microbiota (GM). The GM is a vital modulator of brain function through the gut-brain axis, which acts as the neural pathway between the central and peripheral nervous systems. Dysbiosis of GM plays an essential role in anxiety development because of alterations in the vagus nerve, increased intestinal permeability, and altered breakdown of tryptophan (TRP). The Kynurenine (KYN) pathway plays a crucial role in the pathogenesis of anxiety disorders, primarily through its neuroprotective (KYNA) and neurotoxic (QUIN) metabolites. Higher ratios of KYNA/QUIN result in neuroprotection, whereas higher KYN/TRP ratios indicate increased QUIN production causing neuroinflammation. Studies on germ-free models exhibit higher plasma TRP levels, which interrupt the metabolic balance of TRP-derived compounds, thus causing brain impairment. A key issue in anxiety disorders is the dysregulation of GM, which disrupts TRP metabolism and neuroinflammatory pathways, however, remains poorly understood. Hence, the proper understanding of these mechanisms is crucial for future therapeutic advancements. Here, we highlight the significance of the TRP-KYN pathway and the potential of modulating KYN pathway enzymes, such as kynurenine aminotransferases (KATs), to adjust KYNA levels and restore neurotransmitter balance. It further discusses new therapeutic methods with a particular focus on probiotics that may restore GM and modulate TRP metabolism. Advancing our understanding of the intricate relationship between GM and anxiety disorders may facilitate novel, microbiota-targeted interventions. This ultimately contributes to precision medicine approaches in mental health care, thereby enhancing treatment efficacy and patient outcomes.
Collapse
Affiliation(s)
- Garry Hunjan
- Department of Pharmacy Practice, ISF College of Pharmacy, Moga, 142001, Punjab, India
| | - Shiv Shankar Shah
- Krupanidhi College of Pharmacy, Carmelaram Gunjur Road, Hobli, off Sarjapur Road, Varthur, Bengaluru, 560035, Karnataka, India
| | - Sourabh Kosey
- Department of Pharmacy Practice, ISF College of Pharmacy, Moga, 142001, Punjab, India
| | - Khadga Raj Aran
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, 142001, Punjab, India.
| |
Collapse
|
2
|
Varadan AC, Grasis JA. Filamentous bacteriophage M13 induces proinflammatory responses in intestinal epithelial cells. Infect Immun 2025; 93:e0061824. [PMID: 40208028 PMCID: PMC12070739 DOI: 10.1128/iai.00618-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Accepted: 02/24/2025] [Indexed: 04/11/2025] Open
Abstract
Bacteriophages are the dominant members of the human enteric virome and can shape bacterial communities in the gut; however, our understanding of how they directly impact health and disease is limited. Previous studies have shown that specific bacteriophage populations are expanded in patients with Crohn's disease (CD) and ulcerative colitis (UC), suggesting that fluctuations in the enteric virome may contribute to intestinal inflammation. Based on these studies, we hypothesized that a high bacteriophage burden directly induces intestinal epithelial responses. We found that filamentous bacteriophages M13 and Fd induced dose-dependent IL-8 expression in the human intestinal epithelial cell line HT-29 to a greater degree than their lytic counterparts, T4 and ϕX174. We also found that M13, but not Fd, reduced bacterial internalization in HT-29 cells. This led us to investigate the mechanism underlying M13-mediated inhibition of bacterial internalization by examining the antiviral and antimicrobial responses in these cells. M13 upregulated type I and III IFN expressions and augmented short-chain fatty acid (SCFA)-mediated LL-37 expression in HT-29 cells. Taken together, our data establish that filamentous bacteriophages directly affect human intestinal epithelial cells. These results provide new insights into the complex interactions between bacteriophages and the intestinal mucosa, which may underlie disease pathogenesis.
Collapse
Affiliation(s)
- Ambarish C. Varadan
- Department of Molecular and Cellular Biology, University of California, Merced, California, USA
- Quantitative and Systems Biology Graduate Group, University of California, Merced, California, USA
| | - Juris A. Grasis
- Department of Molecular and Cellular Biology, University of California, Merced, California, USA
- Quantitative and Systems Biology Graduate Group, University of California, Merced, California, USA
| |
Collapse
|
3
|
Martínez‐Augustin O, Tena‐Garitaonaindia M, Ceacero‐Heras D, Jiménez‐Ortas Á, Enguix‐Huete JJ, Álvarez‐Mercado AI, Ruiz‐Henares G, Aranda CJ, Gámez‐Belmonte R, Sánchez de Medina F. Macronutrients as Regulators of Intestinal Epithelial Permeability: Where Do We Stand? Compr Rev Food Sci Food Saf 2025; 24:e70178. [PMID: 40421830 PMCID: PMC12108046 DOI: 10.1111/1541-4337.70178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 03/23/2025] [Accepted: 04/04/2025] [Indexed: 05/28/2025]
Abstract
The intestinal barrier function (IBF) is essential for intestinal homeostasis. Its alterations have been linked to intestinal and systemic disease. Regulation of intestinal permeability is key in the maintenance of the IBF, in which the intestinal epithelium and tight junctions, the mucus layer, sIgA, and antimicrobial peptides are important factors. This review addresses the concept of IBF, focusing on permeability, and summarizes state-of-the-art information on how starvation and macronutrients regulate it. Novel mechanisms regulate intestinal permeability, like its induction by the normal process of nutrient absorption, the contribution of starvation-induced autophagy, or the stimulation of sIgA production by high-protein diets in a T-cell-independent fashion. In addition, observations evidence that starvation and protein restriction increase intestinal permeability, compromising mucin, antimicrobial peptides, and/or intestinal sIgA production. Regarding specific macronutrients, substantial evidence indicates that casein (compared to other protein sources), specific protein-derived peptides and glutamine reinforce IBF. Dietary carbohydrates regulate intestinal permeability in a structure- and composition-dependent fashion; fructose, glucose, and sucrose increase it, while nondigestible oligosaccharides (NDOs) decrease it. Among NDOs, human milk oligosaccharides (HMOs) stand as a promising tool. NODs effects are mediated by intestinal microbiota modulation, production of short-chain fatty acids, and direct interactions with intestinal cells. Finally, evidence supports avoiding high-fat diets for their detrimental effects on IBF. Most studies have been carried out in vitro or in animal models. More information is needed from clinical studies to substantiate beneficial effects and the use of macronutrients in the treatment and prevention of IBF-related diseases.
Collapse
Affiliation(s)
- Olga Martínez‐Augustin
- Department of Biochemistry and Molecular Biology II, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), School of Pharmacy, Insituto de Nutrición y Tecnología de los alimentos José Mataix and Instituto de Investigación Biosanitaria ibs.GRANADAUniversity of GranadaGranadaSpain
| | - Mireia Tena‐Garitaonaindia
- Department of Biochemistry and Molecular Biology II, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), School of Pharmacy, Insituto de Nutrición y Tecnología de los alimentos José Mataix and Instituto de Investigación Biosanitaria ibs.GRANADAUniversity of GranadaGranadaSpain
| | - Diego Ceacero‐Heras
- Department of Biochemistry and Molecular Biology II, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), School of Pharmacy, Insituto de Nutrición y Tecnología de los alimentos José Mataix and Instituto de Investigación Biosanitaria ibs.GRANADAUniversity of GranadaGranadaSpain
| | - Ángela Jiménez‐Ortas
- Department of Biochemistry and Molecular Biology II, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), School of Pharmacy, Insituto de Nutrición y Tecnología de los alimentos José Mataix and Instituto de Investigación Biosanitaria ibs.GRANADAUniversity of GranadaGranadaSpain
| | - Juan J. Enguix‐Huete
- Department of Pharmacology, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), School of Pharmacy, Instituto de Investigación Biosanitaria ibs.GRANADAUniversity of GranadaGranadaSpain
| | - Ana I. Álvarez‐Mercado
- Department of Pharmacology, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), School of Pharmacy, Instituto de Investigación Biosanitaria ibs.GRANADAUniversity of GranadaGranadaSpain
| | - Guillermo Ruiz‐Henares
- Department of Pharmacology, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), School of Pharmacy, Instituto de Investigación Biosanitaria ibs.GRANADAUniversity of GranadaGranadaSpain
| | - Carlos J. Aranda
- Allergy Research Group, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina‐ IBIMA Plataforma BIONANDRICORS “Enfermedades inflamatorias”MálagaSpain
| | - Reyes Gámez‐Belmonte
- Department of Pharmacology, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), School of Pharmacy, Instituto de Investigación Biosanitaria ibs.GRANADAUniversity of GranadaGranadaSpain
- Department of Medicine 1University of Erlangen‐NurembergErlangenGermany
| | - Fermín Sánchez de Medina
- Department of Pharmacology, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), School of Pharmacy, Instituto de Investigación Biosanitaria ibs.GRANADAUniversity of GranadaGranadaSpain
| |
Collapse
|
4
|
Wang W, Wu D, Liu J, Yang DA. Potential protective role of Lycium ruthenicum Murray polysaccharides against lipopolysaccharide-induced liver injury via mitochondrial biogenesis. Int J Biol Macromol 2025; 306:141365. [PMID: 39993693 DOI: 10.1016/j.ijbiomac.2025.141365] [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: 01/27/2024] [Revised: 02/14/2025] [Accepted: 02/19/2025] [Indexed: 02/26/2025]
Abstract
Acute liver injury (ALI), which manifests as abnormal liver function and hepatocyte damage, lacks effective treatment modalities and is associated with a high mortality rate. Recent studies have revealed that hepatoprotection is related to polysaccharide components. In this study, we examined the effect and mechanism of Lycium ruthenicum Murray polysaccharides (LRMP) on liver injury induced by lipopolysaccharide (LPS). Male ICR mice were pre-administered LRMP (100 and 400 mg/kg BW) once daily for 21 days. A single injection of LPS (10 mg/kg BW) was administered on day 21 to induce ALI. The difference between the groups indicated that LRMP supplementation had no adverse effect on body weight. LRMP administration considerably alleviated liver injury, as evidenced by the decreased levels of aspartate transaminase and alanine transaminase, increased levels of albumin, and preservation of liver structural integrity. Moreover, LRMP reduced oxidative stress and inflammatory responses in the liver, maintained mitochondrial structure, regulated mitochondrial apoptotic pathway, and upregulated Sirtuin 1/peroxisome proliferator-activated receptor γ coactivator-1α signalling pathway involved in mitochondrial biogenesis. This study suggests the potential therapeutic application of LRMP in liver-related diseases, which will provide a basis for innovative strategies.
Collapse
Affiliation(s)
- Wenjia Wang
- College of Animal Science, Ningxia University, Yinchuan 750021, China; College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Desheng Wu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Jiaguo Liu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China.
| | - Danchen Aaron Yang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China.
| |
Collapse
|
5
|
Hansell CE, Aneis HA, Kitsios GD, Bain WG, Zhao Y, Suber TL, Evankovich JW, Sharma L, Ramakrishnan SK, Prendergast NT, Hensley MK, Malik S, Petro N, Patel JJ, Nouraie SM, Dela Cruz CS, Zhang Y, McVerry BJ, Shah FA. Glucagon-Like Peptide-1 Is Prognostic of Mortality in Acute Respiratory Failure. Crit Care Explor 2025; 7:e1247. [PMID: 40126931 PMCID: PMC11936568 DOI: 10.1097/cce.0000000000001247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/26/2025] Open
Abstract
OBJECTIVES The incretin hormones glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic peptide (GIP) have therapeutic effects in diabetes mellitus. Prior clinical studies suggest incretins are prognostic of adverse outcomes in critical illness. We investigated whether incretin levels indicate disease severity and clinical outcomes in patients with acute respiratory failure, a common cause of critical illness. DESIGN Retrospective cohort study. SETTING ICUs in UPMC Health Systems hospitals within Western Pennsylvania. PATIENTS Two hundred ninety-seven critically ill adults with acute respiratory failure. INTERVENTIONS None. MEASUREMENTS AND MAIN RESULTS We measured GLP-1 and GIP levels in baseline samples collected at the time of study enrollment. We compared incretin levels across subgroups differing by severity of illness and investigated associations between incretins and markers of systemic host responses and intestinal permeability. In our primary analysis, we tested the association of each incretin level with 90-day mortality by logistic regression in unadjusted analyses and in analyses adjusted for age, Sequential Organ Failure Assessment score, and circulating interleukin-6 levels. GLP-1 levels were higher in nonsurvivors and patients with or at-risk for acute respiratory distress syndrome compared to those intubated for airway protection. GLP-1 levels also positively correlated with systemic immune response biomarkers but not with markers of intestinal permeability. GLP-1 levels positively correlated with mortality in unadjusted (odds ratio, 1.99 [1.55-2.56]; p < 0.01) and adjusted (2.02 [1.23-3.31]; p < 0.01) analyses. GIP levels were not associated with mortality or with host response biomarkers. CONCLUSIONS GLP-1 but not GIP levels were positively associated with systemic inflammation and mortality in critically ill patients with acute respiratory failure. Increased circulating GLP-1 levels may serve as prognostic biomarkers to identify patients who are likely to have worse outcomes.
Collapse
Affiliation(s)
- Cole E. Hansell
- Department of Medicine, UPMC Presbyterian-Shadyside Hospitals, Pittsburgh, PA
| | - Hamam A. Aneis
- Department of Medicine, UPMC McKeesport Hospital, Pittsburgh, PA
| | - Georgios D. Kitsios
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, University of Pittsburgh, Pittsburgh, PA
- Acute Lung Injury and Infection Center of Excellence, University of Pittsburgh, Pittsburgh, PA
- Center for Medicine and the Microbiome, University of Pittsburgh, Pittsburgh, PA
| | - William G. Bain
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, University of Pittsburgh, Pittsburgh, PA
- Acute Lung Injury and Infection Center of Excellence, University of Pittsburgh, Pittsburgh, PA
- Veterans Affairs Pittsburgh Healthcare System, Pulmonary Division, Pittsburgh, PA
| | - Yanwu Zhao
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Tomeka L. Suber
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, University of Pittsburgh, Pittsburgh, PA
- Acute Lung Injury and Infection Center of Excellence, University of Pittsburgh, Pittsburgh, PA
| | - John W. Evankovich
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, University of Pittsburgh, Pittsburgh, PA
- Aging Institute, University of Pittsburgh, Pittsburgh, PA
| | - Lokesh Sharma
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, University of Pittsburgh, Pittsburgh, PA
- Acute Lung Injury and Infection Center of Excellence, University of Pittsburgh, Pittsburgh, PA
| | | | - Niall T. Prendergast
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, University of Pittsburgh, Pittsburgh, PA
- Acute Lung Injury and Infection Center of Excellence, University of Pittsburgh, Pittsburgh, PA
| | - Matthew K. Hensley
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, University of Pittsburgh, Pittsburgh, PA
- Acute Lung Injury and Infection Center of Excellence, University of Pittsburgh, Pittsburgh, PA
| | - Shehryar Malik
- Department of Medicine, UPMC Mercy Hospital, Pittsburgh, PA
| | - Nancy Petro
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, University of Pittsburgh, Pittsburgh, PA
- Acute Lung Injury and Infection Center of Excellence, University of Pittsburgh, Pittsburgh, PA
| | - Jayshil J. Patel
- Division of Pulmonary and Critical Care Medicine, Medical College of Wisconsin, Milwaukee, WI
| | - Seyed Mehdi Nouraie
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, University of Pittsburgh, Pittsburgh, PA
- Acute Lung Injury and Infection Center of Excellence, University of Pittsburgh, Pittsburgh, PA
| | - Charles S. Dela Cruz
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, University of Pittsburgh, Pittsburgh, PA
- Acute Lung Injury and Infection Center of Excellence, University of Pittsburgh, Pittsburgh, PA
- Veterans Affairs Pittsburgh Healthcare System, Pulmonary Division, Pittsburgh, PA
| | - Yingze Zhang
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, University of Pittsburgh, Pittsburgh, PA
- Acute Lung Injury and Infection Center of Excellence, University of Pittsburgh, Pittsburgh, PA
| | - Bryan J. McVerry
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, University of Pittsburgh, Pittsburgh, PA
- Acute Lung Injury and Infection Center of Excellence, University of Pittsburgh, Pittsburgh, PA
| | - Faraaz A. Shah
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, University of Pittsburgh, Pittsburgh, PA
- Acute Lung Injury and Infection Center of Excellence, University of Pittsburgh, Pittsburgh, PA
- Veterans Affairs Pittsburgh Healthcare System, Pulmonary Division, Pittsburgh, PA
| |
Collapse
|
6
|
Abbas Z, Tong Y, Zhang J, Sammad A, Wang J, Ahmad B, Wei X, Si D, Zhang R. Transcriptomics and microbiome insights reveal the protective mechanism of mulberry-derived postbiotics against inflammation in LPS-induced mice. Front Immunol 2025; 16:1536694. [PMID: 40040706 PMCID: PMC11876837 DOI: 10.3389/fimmu.2025.1536694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Accepted: 01/28/2025] [Indexed: 03/06/2025] Open
Abstract
Background Natural food-derived bioactive compounds have garnered increasing attention for their potential to modulate immune responses and promote gut health. In particular, compounds like mulberry-derived postbiotics (MDP) may offer novel therapeutic strategies to address inflammation, a key driver of many metabolic disorders. Methodology This study examines the protective effects of MDP against inflammation in LPS-induced mice, using transcriptomic and microbiome analyses to explore underlying mechanisms. Results MDP pretreatment alleviates LPSinduced villous atrophy and intestinal barrier damage, promoting recovery of intestinal morphology. Transcriptomic profiling revealed significant changes in gene expression, with 983 upregulated and 1220 downregulated genes in the NC vs LPS comparison, and 380 upregulated and 204 downregulated genes in the LPS vs LPS+MDP comparison. Enrichment analysis using GO and KEGG pathways revealed significant associations with transcriptional regulatory activity, and the NOD-like receptor signaling pathway among the differentially expressed genes. Protein-protein interaction analysis identified key genes involved in inflammation and immune regulation, with hub genes like IL6, CXCL10, and MYD88 in the LPS group and CD74, CIITA, and H2-AB1 in the MDP-treated group. Conclusion Microbiome analysis suggested MDP may also influence gut microbiota composition, supporting systemic immune regulation. These findings highlight MDP's potential as a food additive for immune modulation and gut health.
Collapse
Affiliation(s)
- Zaheer Abbas
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Yucui Tong
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Jing Zhang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Abdul Sammad
- Key Laboratory of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Junyong Wang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Baseer Ahmad
- Faculty of Veterinary and Animal Science, Muhammad Nawaz Sharif University of Agriculture, Multan, Pakistan
| | - Xubiao Wei
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Dayong Si
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Rijun Zhang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| |
Collapse
|
7
|
Kalaga P, Ray SK. Mental Health Disorders Due to Gut Microbiome Alteration and NLRP3 Inflammasome Activation After Spinal Cord Injury: Molecular Mechanisms, Promising Treatments, and Aids from Artificial Intelligence. Brain Sci 2025; 15:197. [PMID: 40002529 PMCID: PMC11852823 DOI: 10.3390/brainsci15020197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Revised: 02/06/2025] [Accepted: 02/12/2025] [Indexed: 02/27/2025] Open
Abstract
Aside from its immediate traumatic effects, spinal cord injury (SCI) presents multiple secondary complications that can be harmful to those who have been affected by SCI. Among these secondary effects, gut dysbiosis (GD) and the activation of the NOD (nucleotide-binding oligomerization domain) like receptor-family pyrin-domain-containing three (NLRP3) inflammasome are of special interest for their roles in impacting mental health. Studies have found that the state of the gut microbiome is thrown into disarray after SCI, providing a chance for GD to occur. Metabolites such as short-chain fatty acids (SCFAs) and a variety of neurotransmitters produced by the gut microbiome are hampered by GD. This disrupts healthy cognitive processes and opens the door for SCI patients to be impacted by mental health disorders. Additionally, some studies have found an increased presence and activation of the NLRP3 inflammasome and its respective parts in SCI patients. Preclinical and clinical studies have shown that NLRP3 inflammasome plays a key role in the maturation of pro-inflammatory cytokines that can initiate and eventually aggravate mental health disorders after SCI. In addition to the mechanisms of GD and the NLRP3 inflammasome in intensifying mental health disorders after SCI, this review article further focuses on three promising treatments: fecal microbiome transplants, phytochemicals, and melatonin. Studies have found these treatments to be effective in combating the pathogenic mechanisms of GD and NLRP3 inflammasome, as well as alleviating the symptoms these complications may have on mental health. Another area of focus of this review article is exploring how artificial intelligence (AI) can be used to support treatments. AI models have already been developed to track changes in the gut microbiome, simulate drug-gut interactions, and design novel anti-NLRP3 inflammasome peptides. While these are promising, further research into the applications of AI for the treatment of mental health disorders in SCI is needed.
Collapse
Affiliation(s)
| | - Swapan K. Ray
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine, 6439 Garners Ferry Road, Columbia, SC 29209, USA;
| |
Collapse
|
8
|
Beldie LA, Dica CC, Moța M, Pirvu BF, Burticală MA, Mitrea A, Clenciu D, Efrem IC, Vladu BE, Timofticiuc DCP, Roșu MM, Gheonea TC, Amzolini AM, Moța E, Vladu IM. The Interactions Between Diet and Gut Microbiota in Preventing Gestational Diabetes Mellitus: A Narrative Review. Nutrients 2024; 16:4131. [PMID: 39683525 DOI: 10.3390/nu16234131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 11/26/2024] [Accepted: 11/27/2024] [Indexed: 12/18/2024] Open
Abstract
Recent studies have revealed that dysbiosis, defined as alterations in gut microbiota, plays an important role in the development and the progression of many non-communicable diseases, including metabolic disorders, such as type 2 diabetes mellitus and gestational diabetes mellitus (GDM). The high frequency of GDM makes this disorder an important public health issue, which needs to be addressed in order to reduce both the maternal and fetal complications that are frequently associated with this disease. The studies regarding the connections between gut dysbiosis and GDM are still in their early days, with new research continuously emerging. This narrative review seeks to outline the mechanisms through which a healthy diet that protects the gut microbiota is able to prevent the occurrence of GDM, thus providing medical nutritional therapeutic perspectives for the management of GDM.
Collapse
Affiliation(s)
- Luiza-Andreea Beldie
- Department of Diabetes, Nutrition and Metabolic Diseases, County Clinical Emergency Hospital of Craiova, 200642 Craiova, Romania
| | - Cristina-Camelia Dica
- Department of Diabetes, Nutrition and Metabolic Diseases, County Clinical Emergency Hospital of Craiova, 200642 Craiova, Romania
| | - Maria Moța
- Doctoral School, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Bianca-Florentina Pirvu
- Department of Diabetes, Nutrition and Metabolic Diseases, County Clinical Emergency Hospital of Craiova, 200642 Craiova, Romania
| | - Marilena-Alexandra Burticală
- Department of Diabetes, Nutrition and Metabolic Diseases, County Clinical Emergency Hospital of Craiova, 200642 Craiova, Romania
| | - Adina Mitrea
- Department of Diabetes, Nutrition and Metabolic Diseases, Faculty of Medicine, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Diana Clenciu
- Department of Diabetes, Nutrition and Metabolic Diseases, Faculty of Medicine, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Ion Cristian Efrem
- Department of Medical Semiology, Faculty of Dentistry, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Beatrice Elena Vladu
- Faculty of Medicine, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Diana Cristina Protasiewicz Timofticiuc
- Department of Diabetes, Nutrition and Metabolic Diseases, County Clinical Emergency Hospital of Craiova, 200642 Craiova, Romania
- Department of Diabetes, Nutrition and Metabolic Diseases, Faculty of Midwives and Nursing, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Maria Magdalena Roșu
- Department of Diabetes, Nutrition and Metabolic Diseases, Faculty of Midwives and Nursing, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Theodora Claudia Gheonea
- Department of Diabetes, Nutrition and Metabolic Diseases, Faculty of Medicine, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Anca Maria Amzolini
- Department of Medical Semiology, Faculty of Medicine, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Eugen Moța
- Doctoral School, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Ionela Mihaela Vladu
- Department of Diabetes, Nutrition and Metabolic Diseases, Faculty of Medicine, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| |
Collapse
|
9
|
Zhao J, Jia W, Zhang R, Wang X, Zhang L. Improving curcumin bioavailability: Targeted delivery of curcumin and loading systems in intestinal inflammation. Food Res Int 2024; 196:115079. [PMID: 39614566 DOI: 10.1016/j.foodres.2024.115079] [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: 05/23/2024] [Revised: 09/04/2024] [Accepted: 09/10/2024] [Indexed: 12/01/2024]
Abstract
Curcumin is a natural food ingredient and has the potential to alleviate inflammation and combat cancer. The incidence of intestinal inflammation has been increasing and poses a severe risk to human health. Due to low absorption and bioavailability, curcumin's anti-inflammatory ability is ineffective. To improve the bioavailability of curcumin, descriptions of the intestinal barrier, signaling pathways, and transport mechanisms are reviewed. Blocking the signaling pathways lowers the number of inflammatory cytokines produced, which is the primary mechanism by which curcumin relieves inflammatory symptoms. The bioavailability of curcumin is not only related to physicochemical properties but also to the nature of the carrier material. Environmental indicators also have an impact on the improvement of curcumin bioavailability in applications. There is a need to develop multifunctional and more stable nanomaterial targeting systems to improve curcumin bioavailability and achieve better results in nanotechnology research and targeted inflammation therapy.
Collapse
Affiliation(s)
- Junyi Zhao
- School of Food and Biological Engineering, Shaanxi University of Science & Technology, Xi'an 710021, China
| | - Wei Jia
- School of Food and Biological Engineering, Shaanxi University of Science & Technology, Xi'an 710021, China.
| | - Rong Zhang
- School of Food and Biological Engineering, Shaanxi University of Science & Technology, Xi'an 710021, China
| | - Xin Wang
- School of Food and Biological Engineering, Shaanxi University of Science & Technology, Xi'an 710021, China
| | - Li Zhang
- Keyi Sunshine Test, Xi'an 710021, China
| |
Collapse
|
10
|
Radler JB, McBride AR, Saha K, Nighot P, Holmes GM. Regional Heterogeneity in Intestinal Epithelial Barrier Permeability and Mesenteric Perfusion After Thoracic Spinal Cord Injury. Dig Dis Sci 2024; 69:3236-3248. [PMID: 39001959 DOI: 10.1007/s10620-024-08537-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 06/18/2024] [Indexed: 07/15/2024]
Abstract
BACKGROUND Spinal cord injury (SCI) disrupts intestinal barrier function, thereby increasing antigen permeation and leading to poor outcomes. Despite the intestinal tract's anatomic and physiologic heterogeneity, studies following SCI have not comprehensively addressed intestinal pathophysiology with regional specificity. AIMS AND METHODS We used an experimental model of high thoracic SCI to investigate (1) regional mucosal oxidative stress using dihydroethidium labeling; (2) regional paracellular permeability to small- and large-molecular probes via Ussing chamber; (3) regional intestinal tight junction (TJ) protein expression; and (4) hindgut perfusion via the caudal mesenteric artery. RESULTS Dihydroethidium staining was significantly elevated within duodenal mucosa at 3-day post-SCI. Molar flux of [14C]-urea was significantly elevated in duodenum and proximal colon at 3-day post-SCI, while molar flux of [3H]-inulin was significantly elevated only in duodenum at 3-day post-SCI. Barrier permeability was mirrored by a significant increase in the expression of pore-forming TJ protein claudin-2 in duodenum and proximal colon at 3-day post-SCI. Claudin-2 expression remained significantly elevated in proximal colon at 3-week post-SCI. Expression of the barrier-forming TJ protein occludin was significantly reduced in duodenum at 3-day post-SCI. Caudal mesenteric artery flow was unchanged by SCI at 3 days or 3 weeks despite significant reductions in mean arterial pressure. CONCLUSION These data show that T3-SCI provokes elevated mucosal oxidative stress, altered expression of TJ proteins, and elevated intestinal barrier permeability in the proximal intestine. In contrast, mucosal oxidative stress and intestinal barrier permeability were unchanged in the hindgut after SCI. This regional heterogeneity may result from differential sensitivity to reduced mesenteric perfusion, though further studies are required to establish a causal link. Understanding regional differences in intestinal pathophysiology is essential for developing effective treatments and standards of care for individuals with SCI.
Collapse
Affiliation(s)
- Jackson B Radler
- Department of Neural and Behavioral Sciences, Penn State University College of Medicine, 500 University Dr., H109, Hershey, PA, 17033, USA
| | - Amanda R McBride
- Department of Neural and Behavioral Sciences, Penn State University College of Medicine, 500 University Dr., H109, Hershey, PA, 17033, USA
- Cooper Medical School of Rowan University, Camden, NJ, 08103, USA
| | - Kushal Saha
- Division of Gastroenterology and Hepatology, Department of Medicine, Penn State University College of Medicine, Hershey, PA, 17033, USA
| | - Prashant Nighot
- Division of Gastroenterology and Hepatology, Department of Medicine, Penn State University College of Medicine, Hershey, PA, 17033, USA
| | - Gregory M Holmes
- Department of Neural and Behavioral Sciences, Penn State University College of Medicine, 500 University Dr., H109, Hershey, PA, 17033, USA.
| |
Collapse
|
11
|
Choi Y, Park H, Kim J, Lee H, Kim M. Heat Stress Induces Alterations in Gene Expression of Actin Cytoskeleton and Filament of Cellular Components Causing Gut Disruption in Growing-Finishing Pigs. Animals (Basel) 2024; 14:2476. [PMID: 39272260 PMCID: PMC11394201 DOI: 10.3390/ani14172476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 08/17/2024] [Accepted: 08/22/2024] [Indexed: 09/15/2024] Open
Abstract
We aimed to investigate the impact of heat stress (HS) on the expression of tight junction (TJ) proteins and the interaction between genes affecting intestinal barrier function using transcriptomics in the porcine jejunum. Twenty-four barrows (crossbred Yorkshire × Landrace × Duroc; average initial body weight, 56.71 ± 1.74 kg) were placed in different temperatures (normal temperature [NT]; HS) and reared for 56 days. At the end of the experiment, jejunal samples were collected from three pigs per treatment for transcriptome and reverse-transcription quantitative polymerase chain reaction (RT-qPCR) analyses. We identified 43 differentially expressed genes, involving five Kyoto Encyclopedia of Genes and Genomes pathways, eight molecular functions, seven cellular components (CCs), and nine biological processes, using gene ontology enrichment analysis. Genes associated with the actin cytoskeleton, filament-binding pathways, and TJ proteins were selected and analyzed by RT-qPCR. Significant differences in relative mRNA expression showed that downregulated genes in the HS group included ZO1, CLDN1, OCLN, PCK1, and PCK2, whereas ACTG2, DES, MYL9, MYLK, TPM1, TPM2, CNN1, PDLIM3, and PCP4 were upregulated by HS (p < 0.05). These findings indicate that HS in growing-finishing pigs induces depression in gut integrity, which may be related to genes involved in the actin cytoskeleton and filaments of CC.
Collapse
Affiliation(s)
- Yohan Choi
- Swine Science Division, National Institute of Animal Science, Rural Development Administration, Cheonan 31000, Republic of Korea
| | - Hyunju Park
- Swine Science Division, National Institute of Animal Science, Rural Development Administration, Cheonan 31000, Republic of Korea
| | - Joeun Kim
- Swine Science Division, National Institute of Animal Science, Rural Development Administration, Cheonan 31000, Republic of Korea
| | - Hyunseo Lee
- School of Animal Life Convergence Science, Hankyong National University, Anseong 17579, Republic of Korea
| | - Minju Kim
- School of Animal Life Convergence Science, Hankyong National University, Anseong 17579, Republic of Korea
- Institute of Applied Humanimal Science, Hankyong National University, Anseong 17579, Republic of Korea
| |
Collapse
|
12
|
Reale M, Costantini E, Aielli L, Rienzo AD, Biase GD, Stefano AD, Cacciatore I. Exploring the therapeutic potential of cinnamoyl derivatives in attenuating inflammation in lipopolysaccharide-induced Caco-2 cells. Future Med Chem 2024; 16:1395-1411. [PMID: 39190472 PMCID: PMC11352700 DOI: 10.1080/17568919.2024.2351293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 04/29/2024] [Indexed: 08/28/2024] Open
Abstract
Aim: In gastrointestinal (GI) diseases, lipopolysaccharide (LPS) exacerbates gut-barrier dysfunction and inflammation. Cinnamoyl derivatives show potential in mitigating LPS-induced inflammation.Materials & methods: We assessed intestinal epithelial barrier function using Trans-epithelial electrical resistance values and measured inflammatory mediators through real-time PCR and ELISA in Caco-2 cells.Results: LPS treatment increased IL-6, IL-1β, TNF-α, PGE2 and TRL4 expression in Caco-2 cells. Pre-treatment with DM1 (1 or 10 μM) effectively countered LPS-induced TLR4 overexpression and reduced IL-6, IL-1β, TNF-α and PGE2 levels.Conclusion: DM1 holds promise in regulating inflammation and maintaining intestinal integrity by suppressing TLR4 and inflammatory mediators in Caco-2 cells. These findings suggest a potential therapeutic avenue for GI diseases.
Collapse
Affiliation(s)
- Marcella Reale
- Department of Innovative Technologies in Medicine & Dentistry, University “G. d'Annunzio”, Via dei Vestini, 66100, Chieti, Italy
| | - Erica Costantini
- Department of Medicine & Aging Sciences, University “G. d'Annunzio”, Via dei Vestini, 66100, Chieti, Italy
| | - Lisa Aielli
- Department of Innovative Technologies in Medicine & Dentistry, University “G. d'Annunzio”, Via dei Vestini, 66100, Chieti, Italy
| | - Annalisa Di Rienzo
- Department of Pharmacy, University “G. d'Annunzio”, Via dei Vestini, 66100, Chieti, Italy
| | - Giuseppe Di Biase
- Department of Pharmacy, University “G. d'Annunzio”, Via dei Vestini, 66100, Chieti, Italy
| | - Antonio Di Stefano
- Department of Pharmacy, University “G. d'Annunzio”, Via dei Vestini, 66100, Chieti, Italy
| | - Ivana Cacciatore
- Department of Pharmacy, University “G. d'Annunzio”, Via dei Vestini, 66100, Chieti, Italy
| |
Collapse
|
13
|
Chunchai T, Chinchapo T, Sripetchwandee J, Thonusin C, Chattipakorn N, Chattipakorn SC. Lipopolysaccharide exacerbates depressive-like behaviors in obese rats through complement C1q-mediated synaptic elimination by microglia. Acta Physiol (Oxf) 2024; 240:e14130. [PMID: 38462756 DOI: 10.1111/apha.14130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 02/05/2024] [Accepted: 02/28/2024] [Indexed: 03/12/2024]
Abstract
AIM Prolonged high-fat diet (HFD) consumption has been shown to impair cognition and depression. The combined effects of HFD and lipopolysaccharide (LPS) administration on those outcomes have never been thoroughly investigated. This study investigated the effects of LPS, HFD consumption, and a combination of both conditions on microglial dysfunction, microglial morphological alterations, synaptic loss, cognitive dysfunction, and depressive-like behaviors. METHODS Sixty-four male Wistar rats were fed either a normal diet (ND) or HFD for 12 weeks, followed by single dose-subcutaneous injection of either vehicle or LPS. Then, cognitive function and depressive-like behaviors were assessed. Then, rats were euthanized, and the whole brain, hippocampus, and spleen were collected for further investigation, including western blot analysis, qRT-PCR, immunofluorescence staining, and brain metabolome determination. RESULTS HFD-fed rats developed obese characteristics. Both HFD-fed rats with vehicle and ND-fed rats with LPS increased cholesterol and serum LPS levels, which were exacerbated in HFD-fed rats with LPS. HFD consumption, but not LPS injection, caused oxidative stress, blood-brain barrier disruption, and decreased neurogenesis. Both HFD and LPS administration triggered an increase in inflammatory genes on microglia and astrocytes, increased c1q colocalization with microglia, and increased dendritic spine loss, which were exacerbated in the combined conditions. Both HFD and LPS altered neurotransmitters and disrupted brain metabolism. Interestingly, HFD consumption, but not LPS, induced cognitive decline, whereas both conditions individually induced depressive-like behaviors, which were exacerbated in the combined conditions. CONCLUSIONS Our findings suggest that LPS aggravates metabolic disturbances, neuroinflammation, microglial synaptic engulfment, and depressive-like behaviors in obese rats.
Collapse
Affiliation(s)
- Titikorn Chunchai
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Thirathada Chinchapo
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Jirapas Sripetchwandee
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Chanisa Thonusin
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Nipon Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Siriporn C Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
- Department of Oral Biology and Diagnostic Sciences, Faculty of Dentistry, Chiang Mai University, Chiang Mai, Thailand
| |
Collapse
|
14
|
Deng W, Yi P, Xiong Y, Ying J, Lin Y, Dong Y, Wei G, Wang X, Hua F. Gut Metabolites Acting on the Gut-Brain Axis: Regulating the Functional State of Microglia. Aging Dis 2024; 15:480-502. [PMID: 37548933 PMCID: PMC10917527 DOI: 10.14336/ad.2023.0727] [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: 05/25/2023] [Accepted: 07/27/2023] [Indexed: 08/08/2023] Open
Abstract
The gut-brain axis is a communication channel that mediates a complex interplay of intestinal flora with the neural, endocrine, and immune systems, linking gut and brain functions. Gut metabolites, a group of small molecules produced or consumed by biochemical processes in the gut, are involved in central nervous system regulation via the highly interconnected gut-brain axis affecting microglia indirectly by influencing the structure of the gut-brain axis or directly affecting microglia function and activity. Accordingly, pathological changes in the central nervous system are connected with changes in intestinal metabolite levels as well as altered microglia function and activity, which may contribute to the pathological process of each neuroinflammatory condition. Here, we discuss the mechanisms by which gut metabolites, for instance, the bile acids, short-chain fatty acids, and tryptophan metabolites, regulate the structure of each component of the gut-brain axis, and explore the important roles of gut metabolites in the central nervous system from the perspective of microglia. At the same time, we highlight the roles of gut metabolites affecting microglia in the pathogenesis of neurodegenerative diseases and neurodevelopmental disorders. Understanding the relationship between microglia, gut microbiota, neuroinflammation, and neurodevelopmental disorders will help us identify new strategies for treating neuropsychiatric disorders.
Collapse
Affiliation(s)
- Wenze Deng
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China.
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang City, Jiangxi, China.
| | - Pengcheng Yi
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China.
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang City, Jiangxi, China.
| | - Yanhong Xiong
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China.
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang City, Jiangxi, China.
| | - Jun Ying
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China.
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang City, Jiangxi, China.
| | - Yue Lin
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China.
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang City, Jiangxi, China.
| | - Yao Dong
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China.
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang City, Jiangxi, China.
| | - Gen Wei
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China.
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang City, Jiangxi, China.
| | - Xifeng Wang
- Department of Anesthesiology, the First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China.
| | - Fuzhou Hua
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China.
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang City, Jiangxi, China.
| |
Collapse
|
15
|
Reisinger N, Doupovec B, Czabany T, Van Immerseel F, Croubels S, Antonissen G. Endotoxin Translocation Is Increased in Broiler Chickens Fed a Fusarium Mycotoxin-Contaminated Diet. Toxins (Basel) 2024; 16:167. [PMID: 38668592 PMCID: PMC11053883 DOI: 10.3390/toxins16040167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 03/14/2024] [Accepted: 03/19/2024] [Indexed: 04/29/2024] Open
Abstract
Broiler chickens in livestock production face numerous challenges that can impact their health and welfare, including mycotoxin contamination and heat stress. In this study, we aimed to investigate the combined effects of two mycotoxins, deoxynivalenol (DON) and fumonisins (FBs), along with short-term heat stress conditions, on broiler gut health and endotoxin translocation. An experiment was conducted to assess the impacts of mycotoxin exposure on broilers, focusing on intestinal endotoxin activity, gene expression related to gut barrier function and inflammation, and the plasma concentration of the endotoxin marker 3-OH C14:0 either at thermoneutral conditions or short-term heat stress conditions. Independently of heat stress, broilers fed DON-contaminated diets exhibited reduced body weight gain during the starter phase (Day 1-12) compared to the control group, while broilers fed FB-contaminated diets experienced decreased body weight gain throughout the entire trial period (Day 1-24). Furthermore, under thermoneutral conditions, broilers fed DON-contaminated diets showed an increase in 3-OH C14:0 concentration in the plasma. Moreover, under heat stress conditions, the expression of genes related to gut barrier function (Claudin 5, Zonulin 1 and 2) and inflammation (Toll-like receptor 4, Interleukin-1 beta, Interleukin-6) was significantly affected by diets contaminated with mycotoxins, depending on the gut segment. This effect was particularly prominent in broilers fed diets contaminated with FBs. Notably, the plasma concentration of 3-OH C14:0 increased in broilers exposed to both DON- and FB-contaminated diets under heat stress conditions. These findings shed light on the intricate interactions between mycotoxins, heat stress, gut health, and endotoxin translocation in broiler chickens, highlighting the importance of understanding these interactions for the development of effective management strategies in livestock production to enhance broiler health and welfare.
Collapse
Affiliation(s)
- Nicole Reisinger
- dsm-firmenich Animal Nutrition and Health R&D Center Tulln, Technopark 1, 3430 Tulln, Austria; (B.D.); (T.C.)
| | - Barbara Doupovec
- dsm-firmenich Animal Nutrition and Health R&D Center Tulln, Technopark 1, 3430 Tulln, Austria; (B.D.); (T.C.)
| | - Tibor Czabany
- dsm-firmenich Animal Nutrition and Health R&D Center Tulln, Technopark 1, 3430 Tulln, Austria; (B.D.); (T.C.)
| | - Filip Van Immerseel
- Department of Pathobiology, Pharmacology and Zoological Medicine, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820 Merelbeke, Belgium; (F.V.I.); (S.C.); (G.A.)
| | - Siska Croubels
- Department of Pathobiology, Pharmacology and Zoological Medicine, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820 Merelbeke, Belgium; (F.V.I.); (S.C.); (G.A.)
| | - Gunther Antonissen
- Department of Pathobiology, Pharmacology and Zoological Medicine, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820 Merelbeke, Belgium; (F.V.I.); (S.C.); (G.A.)
- Chair Poultry Health Sciences, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820 Merelbeke, Belgium
| |
Collapse
|
16
|
Nguyen OTP, Misun PM, Hierlemann A, Lohasz C. A Versatile Intestine-on-Chip System for Deciphering the Immunopathogenesis of Inflammatory Bowel Disease. Adv Healthc Mater 2024; 13:e2302454. [PMID: 38253407 PMCID: PMC11468350 DOI: 10.1002/adhm.202302454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 12/21/2023] [Indexed: 01/24/2024]
Abstract
The multifactorial nature of inflammatory bowel disease (IBD) necessitates reliable and practical experimental models to elucidate its etiology and pathogenesis. To model the intestinal microenvironment at the onset of IBD in vitro, it is important to incorporate relevant cellular and noncellular components before inducing stepwise pathogenic developments. A novel intestine-on-chip system for investigating multiple aspects of IBD's immunopathogenesis is presented. The system includes an array of tight and polarized barrier models formed from intestinal epithelial cells on an in-vivo-like subepithelial matrix within one week. The dynamic remodeling of the subepithelial matrix by cells or their secretome demonstrates the physiological relevance of the on-chip barrier models. The system design enables introduction of various immune cell types and inflammatory stimuli at specific locations in the same barrier model, which facilitates investigations of the distinct roles of each cell type in intestinal inflammation development. It is showed that inflammatory behavior manifests in an upregulated expression of inflammatory markers and cytokines (TNF-α). The neutralizing effect of the anti-inflammatory antibody Infliximab on levels of TNF-α and its inducible cytokines could be explicitly shown. Overall, an innovative approach to systematically developing a microphysiological system to comprehend immune-system-mediated disorders of IBD and to identify new therapeutic strategies is presented.
Collapse
Affiliation(s)
- Oanh T. P. Nguyen
- Bio Engineering LaboratoryDepartment of Biosystems Science and EngineeringETH ZurichKlingelbergstrasse 48BaselCH‐4056Switzerland
| | - Patrick M. Misun
- Bio Engineering LaboratoryDepartment of Biosystems Science and EngineeringETH ZurichKlingelbergstrasse 48BaselCH‐4056Switzerland
| | - Andreas Hierlemann
- Bio Engineering LaboratoryDepartment of Biosystems Science and EngineeringETH ZurichKlingelbergstrasse 48BaselCH‐4056Switzerland
| | - Christian Lohasz
- Bio Engineering LaboratoryDepartment of Biosystems Science and EngineeringETH ZurichKlingelbergstrasse 48BaselCH‐4056Switzerland
| |
Collapse
|
17
|
Zhou Y, Huang Y, Yang C, Zang X, Deng H, Liu J, Zhao E, Tian T, Pan L, Xue X. The pathways and the mechanisms by which Cryptococcus enters the brain. Mycology 2024; 15:345-359. [PMID: 39247889 PMCID: PMC11376299 DOI: 10.1080/21501203.2023.2295409] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 12/11/2023] [Indexed: 09/10/2024] Open
Abstract
Generally, Cryptococcus initially infects the respiratory tract, but can spread, eventually crossing the blood-brain barrier (BBB) and causing meningitis or meningoencephalitis. Specifically, Cryptococcus invades the vascular endothelial cells of the BBB, from which it enters the brain. The main mechanisms through which Cryptococcus crosses the BBB are transcellular traversal, the paracellular pathway, and via Trojan horse. In this paper, the mechanisms by which Cryptococcus crosses the BBB were explained in detail. In addition to pathways of entry to the brain, this paper presents a discussion on some rare cryptococcal infections and provides some insights for future research directions.
Collapse
Affiliation(s)
- Yangyu Zhou
- Department of Respiratory and Critical Care, Emergency and Critical Care Medical Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Yemei Huang
- Department of Respiratory and Critical Care, Emergency and Critical Care Medical Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Chen Yang
- Department of Respiratory and Critical Care, Weifang Medical College, Weifang, China
| | - Xuelei Zang
- Department of Respiratory and Critical Care, Emergency and Critical Care Medical Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Hengyu Deng
- Department of Laboratory Medicine, Chinese PLA General Hospital, the First Medical Centre, Beijing, China
| | - Jing Liu
- Department of Laboratory Medicine, Chinese PLA General Hospital, the First Medical Centre, Beijing, China
| | - Enqi Zhao
- Department of Respiratory and Critical Care, Emergency and Critical Care Medical Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Tingyue Tian
- Department of Respiratory and Critical Care, Emergency and Critical Care Medical Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Lei Pan
- Department of Respiratory and Critical Care, Emergency and Critical Care Medical Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Xinying Xue
- Department of Respiratory and Critical Care, Emergency and Critical Care Medical Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
- Department of Laboratory Medicine, Chinese PLA General Hospital, the First Medical Centre, Beijing, China
| |
Collapse
|
18
|
Feng H, Xiong J, Liang S, Wang Y, Zhu Y, Hou Q, Yang X, Yang X. Fecal virus transplantation has more moderate effect than fecal microbiota transplantation on changing gut microbial structure in broiler chickens. Poult Sci 2024; 103:103282. [PMID: 38147728 PMCID: PMC10874774 DOI: 10.1016/j.psj.2023.103282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 11/07/2023] [Accepted: 11/12/2023] [Indexed: 12/28/2023] Open
Abstract
Growing evidence of fecal microbiota transplantation (FMT) and fecal virus transplantation (FVT) provides a possibility to regulate animal health, whereas little is known about the impact of the 2 methods. This study aimed to investigate the effects of gut microbes on jejunal function in healthy broiler chickens, with the objective of establishing a theoretical basis for the application of FMT and FVT. Cecal feces from 28-day-old AA broilers were collected to prepare gavage juice for FMT and FVT. FMT for Group FM, FVT for group FV and PBS gavage for group CON, continuously treated for 6 days start at 5-day-old chicks. Samples were collected at d 11 and d 21. The results showed that the treatment d 2 and the overall fecal score in treatment groups were significantly lower than CON group (P < 0.05). The jejunum morphology showed that FMT increased crypt depth, decreased villus height, V/C (P < 0.05) and FVT increased villus height (P < 0.05) at d 11. At d 21, villus height and crypt depth significantly higher (P < 0.05) in group FM and group FV. The expression of Claudin1, Occludin, ZO2, and Muc2 in the FV group was significantly increased (P < 0.05) at 11-day-old. FMT increased the secretion of sIgA at 11-day-old, and this influence lasted up to 21-day-old (P < 0.05). At 11-day-old, the expression of b0+AT of basic amino acid transport carrier and chymotrypsin activity (P < 0.05) had a significant correlation. At 21 d of age, FVT significantly increased the expression of PepT1 and SGLT1 (P < 0.05). At 11-day-old, FM group showed significantly higher faith pd index (P = 0.004) and Shannon index (P = 0.037), and separated from FV and CON according to PCoA. Among differentiating bacteria, Bacteroides significantly enriched (P < 0.05) in group FM, which positively correlated with the expression of ZO2, Muc2, Occludin, and Claudin1; R_Ruminococcus, L_Ruminococcus, Butyricicoccuss significantly enriched (P < 0.05) in group CON, which significantly higher than processing groups, R_Ruminococcus and L_Ruminococcus negatively correlated with the expression of Occludin (P < 0.05), and R_Ruminococcus, Butyricicoccus negatively correlated with the expression of Claudin1 (P < 0.05). At 21-day-old, PCoA based on Bray-Curtis shows that microbes taxa of 3 groups are isolated with each other and treatment groups were significant different with CON group based on Unweighted UniFrac and weighted UniFrac. The expression of PepT1 was significantly negatively (P < 0.05) correlated with Ruminococcus, and the expression of sIgA was significantly negatively (P < 0.05) correlated with Parabacteroides. In conclusion, FMT regulated intestinal flora rapidly, while it had little effect on intestinal function and a higher potential damaging risk on jejunal. FVT regulated intestinal flora structure softer, improved tight junction expression, but the mechanism of action needs further exploration.
Collapse
Affiliation(s)
- Hongyu Feng
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, PR China
| | - Jiaying Xiong
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, PR China
| | - Saisai Liang
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, PR China
| | - Yinlong Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, PR China
| | - Yufei Zhu
- DAYU Bioengeineering (Xi' an) Industrial Development Research Institute. Shaanxi, China; Shanxi Dayu Biological Functions Co., Ltd. Shanxi, China
| | - Qihang Hou
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, PR China
| | - Xiaojun Yang
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, PR China; DAYU Bioengeineering (Xi' an) Industrial Development Research Institute. Shaanxi, China
| | - Xin Yang
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, PR China; DAYU Bioengeineering (Xi' an) Industrial Development Research Institute. Shaanxi, China.
| |
Collapse
|
19
|
Song Q, Zheng Y, Zhong G, Wang S, He C, Li M. Application of Nanoparticles in the Diagnosis and Treatment of Colorectal Cancer. Anticancer Agents Med Chem 2024; 24:1305-1326. [PMID: 39129164 PMCID: PMC11497148 DOI: 10.2174/0118715206323900240807110122] [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: 05/15/2024] [Revised: 06/26/2024] [Accepted: 07/12/2024] [Indexed: 08/13/2024]
Abstract
Colorectal cancer is a common malignant tumor with high morbidity and mortality rates, imposing a huge burden on both patients and the healthcare system. Traditional treatments such as surgery, chemotherapy and radiotherapy have limitations, so finding more effective diagnostic and therapeutic tools is critical to improving the survival and quality of life of colorectal cancer patients. While current tumor targeting research mainly focuses on exploring the function and mechanism of molecular targets and screening for excellent drug targets, it is crucial to test the efficacy and mechanism of tumor cell therapy that targets these molecular targets. Selecting the appropriate drug carrier is a key step in effectively targeting tumor cells. In recent years, nanoparticles have gained significant interest as gene carriers in the field of colorectal cancer diagnosis and treatment due to their low toxicity and high protective properties. Nanoparticles, synthesized from natural or polymeric materials, are NM-sized particles that offer advantages such as low toxicity, slow release, and protection of target genes during delivery. By modifying nanoparticles, they can be targeted towards specific cells for efficient and safe targeting of tumor cells. Numerous studies have demonstrated the safety, efficiency, and specificity of nanoparticles in targeting tumor cells, making them a promising gene carrier for experimental and clinical studies. This paper aims to review the current application of nanoparticles in colorectal cancer diagnosis and treatment to provide insights for targeted therapy for colorectal cancer while also highlighting future prospects for nanoparticle development.
Collapse
Affiliation(s)
- Qiuyu Song
- Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Department of Gastroenterology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yifeng Zheng
- Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Department of Gastroenterology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Guoqiang Zhong
- Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Department of Gastroenterology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Shanping Wang
- Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Department of Gastroenterology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Chengcheng He
- Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Department of Gastroenterology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Mingsong Li
- Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Department of Gastroenterology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
20
|
Jia X, Chen Q, Zhang Y, Asakawa T. Multidirectional associations between the gut microbiota and Parkinson's disease, updated information from the perspectives of humoral pathway, cellular immune pathway and neuronal pathway. Front Cell Infect Microbiol 2023; 13:1296713. [PMID: 38173790 PMCID: PMC10762314 DOI: 10.3389/fcimb.2023.1296713] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 11/27/2023] [Indexed: 01/05/2024] Open
Abstract
The human gastrointestinal tract is inhabited by a diverse range of microorganisms, collectively known as the gut microbiota, which form a vast and complex ecosystem. It has been reported that the microbiota-gut-brain axis plays a crucial role in regulating host neuroprotective function. Studies have shown that patients with Parkinson's disease (PD) have dysbiosis of the gut microbiota, and experiments involving germ-free mice and fecal microbiota transplantation from PD patients have revealed the pathogenic role of the gut microbiota in PD. Interventions targeting the gut microbiota in PD, including the use of prebiotics, probiotics, and fecal microbiota transplantation, have also shown efficacy in treating PD. However, the causal relationship between the gut microbiota and Parkinson's disease remains intricate. This study reviewed the association between the microbiota-gut-brain axis and PD from the perspectives of humoral pathway, cellular immune pathway and neuronal pathway. We found that the interactions among gut microbiota and PD are very complex, which should be "multidirectional", rather than conventionally regarded "bidirectional". To realize application of the gut microbiota-related mechanisms in the clinical setting, we propose several problems which should be addressed in the future study.
Collapse
Affiliation(s)
- Xiaokang Jia
- School of Traditional Chinese Medicine, Hainan Medical University, Haikou, Hainan, China
| | - Qiliang Chen
- School of Basic Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Yuanyuan Zhang
- Department of Acupuncture and Moxibustion, The Affiliated Traditional Chinese Medicine (TCM) Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Tetsuya Asakawa
- Institute of Neurology, National Clinical Research Center for Infectious Diseases, the Third People’s Hospital of Shenzhen, Shenzhen, Guangdong, China
| |
Collapse
|
21
|
Niu X, Hu C, Chen S, Wen J, Liu X, Yong Y, Yu Z, Ma X, Li C, Warda M, Abd El-Aty AM, Gooneratne R, Ju X. Chitosan-gentamicin conjugate attenuates heat stress-induced intestinal barrier injury via the TLR4/STAT6/MYLK signaling pathway: In vitro and in vivo studies. Carbohydr Polym 2023; 321:121279. [PMID: 37739521 DOI: 10.1016/j.carbpol.2023.121279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 08/06/2023] [Accepted: 08/08/2023] [Indexed: 09/02/2023]
Abstract
Heat stress (HS) has a negative impact on animal health. A modified chitosan-gentamicin conjugate (CS-GT) was prepared to investigate its potential protective effects and mechanism of action on heat stress-induced intestinal mucosa injury in IPEC-J2 cells and mouse 3D intestinal organs in a mouse model. CS-GT significantly (P < 0.01) reversed the decline in transmembrane resistance and increased the FITC-dextran permeability of the IPEC-J2 monolayer fusion epithelium caused by heat stress. Heat stress decreased the expression of the tight binding proteins occludin, claudin1, and claudin2. However, pretreatment with CS-GT significantly increased (P < 0.01) the expression of these tight binding proteins. Mechanistically, CS-GT inhibited the activation of the TLR4/STAT6/MYLK signaling pathway induced by heat stress. Molecular docking showed that CS-GT can bind effectively with TLR4. In conclusion, CS-GT alleviates heat stress-induced intestinal mucosal damage both in vitro and in vivo. This effect is mediated, at least partly, by the inhibition of the TLR4/STAT6/MYLK signaling pathway and upregulation of tight junction proteins. These findings suggest that CS-GT may have therapeutic potential in the prevention and treatment of heat stress-related intestinal injury.
Collapse
Affiliation(s)
- Xueting Niu
- College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang 524088, China; Marine Medical Research and Development Centre, Shenzheng Institute of Guangdong Ocean University, Shenzheng 518120, China
| | - Canying Hu
- College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang 524088, China; Marine Medical Research and Development Centre, Shenzheng Institute of Guangdong Ocean University, Shenzheng 518120, China
| | - Shengwei Chen
- College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang 524088, China; Marine Medical Research and Development Centre, Shenzheng Institute of Guangdong Ocean University, Shenzheng 518120, China
| | - Jiaying Wen
- College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang 524088, China; Marine Medical Research and Development Centre, Shenzheng Institute of Guangdong Ocean University, Shenzheng 518120, China
| | - Xiaoxi Liu
- College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang 524088, China
| | - Yanhong Yong
- College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang 524088, China
| | - Zhichao Yu
- College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang 524088, China
| | - Xingbin Ma
- College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang 524088, China
| | - Chengpeng Li
- College of Chemistry and Environment Science, Guangdong Ocean University, Zhanjiang 524088, China
| | - Mohamad Warda
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Cairo University, Giza 12211, Egypt
| | - A M Abd El-Aty
- Department of Pharmacology, Faculty of Veterinary Medicine, Cairo University, 12211 Giza, Egypt; Department of Medical Pharmacology, Medical Faculty, Ataturk University, Erzurum 25240, Turkey
| | - Ravi Gooneratne
- Faculty of Agriculture and Life Sciences, Lincoln University, Lincoln 7647, New Zealand
| | - Xianghong Ju
- College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang 524088, China; Marine Medical Research and Development Centre, Shenzheng Institute of Guangdong Ocean University, Shenzheng 518120, China.
| |
Collapse
|
22
|
Wang Y, Xu Q, Meng M, Chang G, Ma N, Shen X. Butyrate Protects against γ-d-Glutamyl- meso-diaminopimelic Acid-Induced Inflammatory Response and Tight Junction Disruption through Histone Deacetylase 3 Inhibition in Bovine Mammary Epithelial Cells. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:14638-14648. [PMID: 37767922 DOI: 10.1021/acs.jafc.3c04417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/29/2023]
Abstract
The present study was conducted to evaluate the regulatory actions and underlying mechanisms of butyrate on the inflammatory response and tight junction (TJ) disruption in bovine mammary epithelial cells (BMECs). Results showed that butyrate declined histone deacetylase 3 (HDAC3) expression, blocked NF-κB activation, and thus suppressed inflammatory cytokine production in γ-d-glutamyl-meso-diaminopimelic acid (iE-DAP)-triggered BMECs. Butyrate also depressed the protein abundance of myosin light chain kinase (MLCK), elevated the expression of TJ proteins, and restored the cellular distribution of TJ proteins and the barrier function of epithelial cells. HDAC3 overexpression abolished the protective effects of butyrate. In conclusion, butyrate alleviated the iE-DAP-induced inflammatory response and TJ injury by blocking NF-κB activation and decreasing inflammatory cytokine production and MLCK expression in a HDAC3-dependent manner. Our finding provides a mechanistic basis for further exploring the regulatory effects of butyrate on the mammary inflammatory response.
Collapse
Affiliation(s)
- Yan Wang
- College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Qianqian Xu
- College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China
| | - Meijuan Meng
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Guangjun Chang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Nana Ma
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Xiangzhen Shen
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| |
Collapse
|
23
|
Mázala-de-Oliveira T, Silva BT, Campello-Costa P, Carvalho VF. The Role of the Adrenal-Gut-Brain Axis on Comorbid Depressive Disorder Development in Diabetes. Biomolecules 2023; 13:1504. [PMID: 37892186 PMCID: PMC10604999 DOI: 10.3390/biom13101504] [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: 07/04/2023] [Revised: 08/15/2023] [Accepted: 08/26/2023] [Indexed: 10/29/2023] Open
Abstract
Diabetic patients are more affected by depression than non-diabetics, and this is related to greater treatment resistance and associated with poorer outcomes. This increase in the prevalence of depression in diabetics is also related to hyperglycemia and hypercortisolism. In diabetics, the hyperactivity of the HPA axis occurs in parallel to gut dysbiosis, weakness of the intestinal permeability barrier, and high bacterial-product translocation into the bloodstream. Diabetes also induces an increase in the permeability of the blood-brain barrier (BBB) and Toll-like receptor 4 (TLR4) expression in the hippocampus. Furthermore, lipopolysaccharide (LPS)-induced depression behaviors and neuroinflammation are exacerbated in diabetic mice. In this context, we propose here that hypercortisolism, in association with gut dysbiosis, leads to an exacerbation of hippocampal neuroinflammation, glutamatergic transmission, and neuronal apoptosis, leading to the development and aggravation of depression and to resistance to treatment of this mood disorder in diabetic patients.
Collapse
Affiliation(s)
- Thalita Mázala-de-Oliveira
- Laboratório de Inflamação, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21040-360, Brazil; (T.M.-d.-O.); (B.T.S.)
| | - Bruna Teixeira Silva
- Laboratório de Inflamação, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21040-360, Brazil; (T.M.-d.-O.); (B.T.S.)
- Programa de Pós-Graduação em Neurociências, Instituto de Biologia, Universidade Federal Fluminense, Niterói 24210-201, Brazil;
| | - Paula Campello-Costa
- Programa de Pós-Graduação em Neurociências, Instituto de Biologia, Universidade Federal Fluminense, Niterói 24210-201, Brazil;
| | - Vinicius Frias Carvalho
- Laboratório de Inflamação, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21040-360, Brazil; (T.M.-d.-O.); (B.T.S.)
- Programa de Pós-Graduação em Neurociências, Instituto de Biologia, Universidade Federal Fluminense, Niterói 24210-201, Brazil;
- Laboratório de Inflamação, Instituto Nacional de Ciência e Tecnologia em Neuroimunomodulação—INCT-NIM, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21040-360, Brazil
| |
Collapse
|
24
|
Wang YM, Abdullah S, Luebbering N, Langenberg L, Duell A, Lake K, Lane A, Hils B, Vazquez Silva O, Trapp M, Nalapareddy K, Koo J, Denson LA, Jodele S, Haslam DB, Faubion WA, Davies SM, Khandelwal P. Intestinal permeability in patients undergoing stem cell transplantation correlates with systemic acute phase responses and dysbiosis. Blood Adv 2023; 7:5137-5151. [PMID: 37083597 PMCID: PMC10480541 DOI: 10.1182/bloodadvances.2023009960] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 04/05/2023] [Accepted: 04/05/2023] [Indexed: 04/22/2023] Open
Abstract
Intestinal permeability may correlate with adverse outcomes during hematopoietic stem cell transplantation (HSCT), but longitudinal quantification with traditional oral mannitol and lactulose is not feasible in HSCT recipients because of mucositis and diarrhea. A modified lactulose:rhamnose (LR) assay is validated in children with environmental enteritis. Our study objective was to quantify peri-HSCT intestinal permeability changes using the modified LR assay. The LR assay was administered before transplant, at day +7 and +30 to 80 pediatric and young adult patients who received allogeneic HSCT. Lactulose and rhamnose were detected using urine mass spectrometry and expressed as an L:R ratio. Metagenomic shotgun sequencing of stool for microbiome analyses and enzyme-linked immunosorbent assay analyses of plasma lipopolysaccharide binding protein (LBP), ST2, REG3α, claudin1, occludin, and intestinal alkaline phosphatase were performed at the same timepoints. L:R ratios were increased at day +7 but returned to baseline at day +30 in most patients (P = .014). Conditioning regimen intensity did not affect the trajectory of L:R (P = .39). Baseline L:R ratios did not vary with diagnosis. L:R correlated with LBP levels (r2 = 0.208; P = .0014). High L:R ratios were associated with lower microbiome diversity (P = .035), loss of anaerobic organisms (P = .020), and higher plasma LBP (P = .0014). No adverse gastrointestinal effects occurred because of LR. Intestinal permeability as measured through L:R ratios after allogeneic HSCT correlates with intestinal dysbiosis and elevated plasma LBP. The LR assay is well-tolerated and may identify transplant recipients who are more likely to experience adverse outcomes.
Collapse
Affiliation(s)
- YunZu Michele Wang
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
- University of Cincinnati College of Medicine, Cincinnati, OH
| | - Sheyar Abdullah
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
| | - Nathan Luebbering
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
| | - Lucille Langenberg
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
| | - Alexandra Duell
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
| | - Kelly Lake
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
| | - Adam Lane
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
- University of Cincinnati College of Medicine, Cincinnati, OH
| | - Brian Hils
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
| | - Ormarie Vazquez Silva
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
| | - Monica Trapp
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
| | - Kodandaramireddy Nalapareddy
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
| | - Jane Koo
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
- University of Cincinnati College of Medicine, Cincinnati, OH
| | - Lee A. Denson
- University of Cincinnati College of Medicine, Cincinnati, OH
- Department of Gastroenterology, Hepatology and Nutrition, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
| | - Sonata Jodele
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
- University of Cincinnati College of Medicine, Cincinnati, OH
| | - David B. Haslam
- University of Cincinnati College of Medicine, Cincinnati, OH
- Division of Infectious Diseases, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
| | | | - Stella M. Davies
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
- University of Cincinnati College of Medicine, Cincinnati, OH
| | - Pooja Khandelwal
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
- University of Cincinnati College of Medicine, Cincinnati, OH
| |
Collapse
|
25
|
Hamamah S, Amin A, Al-Kassir AL, Chuang J, Covasa M. Dietary Fat Modulation of Gut Microbiota and Impact on Regulatory Pathways Controlling Food Intake. Nutrients 2023; 15:3365. [PMID: 37571301 PMCID: PMC10421457 DOI: 10.3390/nu15153365] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 07/24/2023] [Accepted: 07/25/2023] [Indexed: 08/13/2023] Open
Abstract
Obesity is a multifactorial disease that continues to increase in prevalence worldwide. Emerging evidence has shown that the development of obesity may be influenced by taxonomic shifts in gut microbiota in response to the consumption of dietary fats. Further, these alterations in gut microbiota have been shown to promote important changes in satiation signals including gut hormones (leptin, ghrelin, GLP-1, peptide YY and CCK) and orexigenic and anorexigenic neuropeptides (AgRP, NPY, POMC, CART) that influence hyperphagia and therefore obesity. In this review, we highlight mechanisms by which gut microbiota can influence these satiation signals both locally in the gastrointestinal tract and via microbiota-gut-brain communication. Then, we describe the effects of dietary interventions and associated changes in gut microbiota on satiety signals through microbiota-dependent mechanisms. Lastly, we present microbiota optimizing therapies including prebiotics, probiotics, synbiotics and weight loss surgery that can help restore beneficial gut microbiota by enhancing satiety signals to reduce hyperphagia and subsequent obesity. Overall, a better understanding of the mechanisms by which dietary fats induce taxonomical shifts in gut microbiota and their impact on satiation signaling pathways will help develop more targeted therapeutic interventions in delaying the onset of obesity and in furthering its treatment.
Collapse
Affiliation(s)
- Sevag Hamamah
- Department of Basic Medical Sciences, Western University of Health Sciences, College of Osteopathic Medicine, Pomona, CA 91766, USA; (S.H.); (A.A.); (A.L.A.-K.); (J.C.)
| | - Arman Amin
- Department of Basic Medical Sciences, Western University of Health Sciences, College of Osteopathic Medicine, Pomona, CA 91766, USA; (S.H.); (A.A.); (A.L.A.-K.); (J.C.)
| | - Abdul Latif Al-Kassir
- Department of Basic Medical Sciences, Western University of Health Sciences, College of Osteopathic Medicine, Pomona, CA 91766, USA; (S.H.); (A.A.); (A.L.A.-K.); (J.C.)
| | - Judith Chuang
- Department of Basic Medical Sciences, Western University of Health Sciences, College of Osteopathic Medicine, Pomona, CA 91766, USA; (S.H.); (A.A.); (A.L.A.-K.); (J.C.)
| | - Mihai Covasa
- Department of Basic Medical Sciences, Western University of Health Sciences, College of Osteopathic Medicine, Pomona, CA 91766, USA; (S.H.); (A.A.); (A.L.A.-K.); (J.C.)
- Department of Biomedical Sciences, College of Medicine and Biological Science, University of Suceava, 720229 Suceava, Romania
| |
Collapse
|
26
|
Wang Y, Ke W, Gan J, Zhu H, Xie X, He G, Liu S, Huang Y, Tang H. MicroRNA-29b-3p promotes intestinal permeability in IBS-D via targeting TRAF3 to regulate the NF-κB-MLCK signaling pathway. PLoS One 2023; 18:e0287597. [PMID: 37428806 PMCID: PMC10332595 DOI: 10.1371/journal.pone.0287597] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 06/08/2023] [Indexed: 07/12/2023] Open
Abstract
Irritable bowel syndrome with predominant diarrhea (IBS-D) is characterized by increased intestinal permeability. Previous studies have shown that the microRNA-29 gene is involved in the regulation of intestinal permeability in patients with IBS-D. NF-κB was proved to play a key role in inflammatory response of intestine and resultant disruption of tight junction integrity, whose activity could be inhibited by TNF Receptor-Associated Factor 3 (TRAF3). However, the exact mechanism that induces increased intestinal permeability in IBS-D patients has not been clarified. In this study, we found that microRNA-29b‑3p (miR-29b-3p) was significantly upregulated, while TRAF3 was decreased and the NF-κB-MLCK pathway was activated within the colonic tissue of IBS-D patients. Subsequently, we confirmed the targeting relationship between miR-29b-3p and TRAF3 through a double-luciferase reporter assay. Lentivirus transfection of NCM460 cells with miR-29b-3p-overexpressing and -silencing vectors demonstrated that the expression of TRAF3 was negatively correlated with the level of miR-29b-3p. The NF-κB/MLCK pathway was activated in the miR-29b-3p-overexpressing group and inhibited to some extent in the miR-29b-3p-silencing group. Results in WT and miR-29 knockout mice showed that miR-29b-3p levels were increased, TRAF3 levels were decreased, and the NF-κB/MLCK signaling was activated in the WT IBS-D group as compared with the WT control group. The protein levels of TRAF3 and TJs in the miR-29b-/- IBS-D group were partially recovered and NF-κB/MLCK pathway indicators were, to a certain extent, decreased as compared with the WT IBS-D group. These results suggested that miR-29b-3p deletion enhances the TRAF3 level in IBS-D mice and alleviates the high intestinal permeability. In brief, through the analysis of intestinal tissue samples from IBS-D patients and miR-29b-/- IBS-D mice, we showed that miR-29b-3p is involved in the pathogenesis of intestinal hyperpermeability in IBS-D via targeting TRAF3 to regulate the NF-κB-MLCK signaling pathway.
Collapse
Affiliation(s)
- Yongfu Wang
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Wei Ke
- The First Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Jianfeng Gan
- The Second Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - He Zhu
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Xiangyu Xie
- The First Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Guodong He
- The First Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Shan Liu
- The First Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Yusheng Huang
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Hongmei Tang
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| |
Collapse
|
27
|
Bifidobacterium breve Bif11 supplementation improves depression-related neurobehavioural and neuroinflammatory changes in the mouse. Neuropharmacology 2023; 229:109480. [PMID: 36868402 DOI: 10.1016/j.neuropharm.2023.109480] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 02/13/2023] [Accepted: 02/23/2023] [Indexed: 03/05/2023]
Abstract
Gut dysbiosis has been closely linked to the onset and progression of several brain-related disorders such as depression. The administration of microbiota-based formulations such as probiotics helps restore healthy gut flora and plays a role in preventing and treating depression-like behavior. Therefore, we evaluated the efficacy of probiotic supplementation using our recently isolated putative probiotic Bifidobacterium breve Bif11 in ameliorating lipopolysaccharide (LPS)-induced depression-like behavior in male Swiss albino mice. Mice were fed orally with B. breve Bif11 (1 × 1010 CFU and 2 × 1010 CFU) for 21 days before being challenged with a single intraperitoneal LPS injection (0.83 mg/kg). Behavioral, biochemical, histological and molecular analysis were done with an emphasis on inflammatory pathways linked to depression-like behavior. Daily supplementation with B. breve Bif11 for 21 days prevented the onset of depression-like behavior induced by LPS injection, besides reducing the levels of inflammatory cytokines such as matrix metalloproteinase-2, c-reactive protein, interleukin-6, tumor necrosis factor-alpha and nuclear factor kappa-light-chain-enhancer of activated B cells. It also prevented the decrease of the brain-derived neurotrophic factor levels and neuronal cell viability in the prefrontal cortex of LPS-treated mice. Furthermore, we observed that gut permeability was reduced, there was an improved short-chain fatty acid profile and reduced gut dysbiosis in the LPS mice fed with B. breve Bif11. Similarly, we observed a decrease in behavioural deficits and restoration of gut permeability in chronic mild stress. Together, these results would help in deciphering the role of probiotics in the management of neurological disorders where depression, anxiety and inflammation are prominent clinical features.
Collapse
|
28
|
Horowitz A, Chanez-Paredes SD, Haest X, Turner JR. Paracellular permeability and tight junction regulation in gut health and disease. Nat Rev Gastroenterol Hepatol 2023:10.1038/s41575-023-00766-3. [PMID: 37186118 PMCID: PMC10127193 DOI: 10.1038/s41575-023-00766-3] [Citation(s) in RCA: 264] [Impact Index Per Article: 132.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/03/2023] [Indexed: 05/17/2023]
Abstract
Epithelial tight junctions define the paracellular permeability of the intestinal barrier. Molecules can cross the tight junctions via two distinct size-selective and charge-selective paracellular pathways: the pore pathway and the leak pathway. These can be distinguished by their selectivities and differential regulation by immune cells. However, permeability increases measured in most studies are secondary to epithelial damage, which allows non-selective flux via the unrestricted pathway. Restoration of increased unrestricted pathway permeability requires mucosal healing. By contrast, tight junction barrier loss can be reversed by targeted interventions. Specific approaches are needed to restore pore pathway or leak pathway permeability increases. Recent studies have used preclinical disease models to demonstrate the potential of pore pathway or leak pathway barrier restoration in disease. In this Review, we focus on the two paracellular flux pathways that are dependent on the tight junction. We discuss the latest evidence that highlights tight junction components, structures and regulatory mechanisms, their impact on gut health and disease, and opportunities for therapeutic intervention.
Collapse
Affiliation(s)
- Arie Horowitz
- UNIROUEN, INSERM U1245, Normandy Centre for Genomic and Personalized Medicine, Normandie University, Rouen, France
| | - Sandra D Chanez-Paredes
- Laboratory of Mucosal Barrier Pathobiology, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Xenia Haest
- Laboratory of Mucosal Barrier Pathobiology, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jerrold R Turner
- Laboratory of Mucosal Barrier Pathobiology, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
29
|
Nighot M, Liao PL, Morris N, McCarthy D, Dharmaprakash V, Ullah Khan I, Dalessio S, Saha K, Ganapathy AS, Wang A, Ding W, Yochum G, Koltun W, Nighot P, Ma T. Long-Term Use of Proton Pump Inhibitors Disrupts Intestinal Tight Junction Barrier and Exaggerates Experimental Colitis. J Crohns Colitis 2023; 17:565-579. [PMID: 36322638 PMCID: PMC10115233 DOI: 10.1093/ecco-jcc/jjac168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Indexed: 12/03/2022]
Abstract
BACKGROUND Proton pump inhibitors [PPIs] are widely used to treat a number of gastro-oesophageal disorders. PPI-induced elevation in intragastric pH may alter gastrointestinal physiology. The tight junctions [TJs] residing at the apical intercellular contacts act as a paracellular barrier. TJ barrier dysfunction is an important pathogenic factor in inflammatory bowel disease [IBD]. Recent studies suggest that PPIs may promote disease flares in IBD patients. The role of PPIs in intestinal permeability is not clear. AIM The aim of the present study was to study the effect of PPIs on the intestinal TJ barrier function. METHODS Human intestinal epithelial cell culture and organoid models and mouse IBD models of dextran sodium sulphate [DSS] and spontaneous enterocolitis in IL-10-/- mice were used to study the role of PPIs in intestinal permeability. RESULTS PPIs increased TJ barrier permeability via an increase in a principal TJ regulator, myosin light chain kinase [MLCK] activity and expression, in a p38 MAPK-dependent manner. The PPI-induced increase in extracellular pH caused MLCK activation via p38 MAPK. Long-term PPI administration in mice exaggerated the increase in intestinal TJ permeability and disease severity in two independent models of DSS colitis and IL-10-/- enterocolitis. The TJ barrier disruption by PPIs was prevented in MLCK-/- mice. Human database studies revealed increased hospitalizations associated with PPI use in IBD patients. CONCLUSIONS Our results suggest that long-term use of PPIs increases intestinal TJ permeability and exaggerates experimental colitis via an increase in MLCK expression and activity.
Collapse
Affiliation(s)
- Meghali Nighot
- Division of Gastroenterology and Hepatology, Department of Medicine, Pennsylvania State College of Medicine, Hershey, PA 17033, USA
| | - Pei-Luan Liao
- Division of Gastroenterology and Hepatology, Department of Medicine, Pennsylvania State College of Medicine, Hershey, PA 17033, USA
| | - Nathan Morris
- Division of Gastroenterology and Hepatology, Department of Medicine, Pennsylvania State College of Medicine, Hershey, PA 17033, USA
| | - Dennis McCarthy
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of New Mexico, Albuquerque, NM 87131, USA
| | - Viszwapriya Dharmaprakash
- Division of Gastroenterology and Hepatology, Department of Medicine, Pennsylvania State College of Medicine, Hershey, PA 17033, USA
| | - Inam Ullah Khan
- Division of Gastroenterology and Hepatology, Department of Medicine, Pennsylvania State College of Medicine, Hershey, PA 17033, USA
| | - Shannon Dalessio
- Division of Gastroenterology and Hepatology, Department of Medicine, Pennsylvania State College of Medicine, Hershey, PA 17033, USA
| | - Kushal Saha
- Division of Gastroenterology and Hepatology, Department of Medicine, Pennsylvania State College of Medicine, Hershey, PA 17033, USA
| | | | - Alexandra Wang
- Division of Gastroenterology and Hepatology, Department of Medicine, Pennsylvania State College of Medicine, Hershey, PA 17033, USA
| | - Wei Ding
- Division of Colon and Rectal Surgery, Department of Surgery, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Gregory Yochum
- Division of Colon and Rectal Surgery, Department of Surgery, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Walter Koltun
- Division of Colon and Rectal Surgery, Department of Surgery, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Prashant Nighot
- Division of Gastroenterology and Hepatology, Department of Medicine, Pennsylvania State College of Medicine, Hershey, PA 17033, USA
| | - Thomas Ma
- Division of Gastroenterology and Hepatology, Department of Medicine, Pennsylvania State College of Medicine, Hershey, PA 17033, USA
| |
Collapse
|
30
|
Wang Y, Li X, Han Z, Meng M, Shi X, Wang L, Chen M, Chang G, Shen X. iE-DAP Induced Inflammatory Response and Tight Junction Disruption in Bovine Mammary Epithelial Cells via NOD1-Dependent NF-κB and MLCK Signaling Pathway. Int J Mol Sci 2023; 24:ijms24076263. [PMID: 37047240 PMCID: PMC10094069 DOI: 10.3390/ijms24076263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 03/19/2023] [Accepted: 03/23/2023] [Indexed: 03/29/2023] Open
Abstract
γ-D-glutamyl-meso-diaminopimelic acid (iE-DAP), a bacterial cell wall component, can trigger an inflammatory response. A mammary inflammatory response causes tight junction (TJ) dysfunction. This study aimed to explore the effects and involved mechanisms of iE-DAP-induced inflammatory response on the TJ integrity in bovine mammary epithelial cells (BMECs). The results showed that iE-DAP-induced inflammatory response and TJ disruption was associated with increased expression levels of inflammatory cytokines and decreased gene expression of ZO-1 and Occludin, as well as a reduction in transepithelial electrical resistance and elevation in paracellular dextran passage. While MLCK inhibitor ML-7 reversed the TJ disruption induced by iE-DAP. NF-κB inhibitor BAY 11-7085 hindered the activation of NF-κB and MLCK signaling pathways, the inflammatory response and TJ disruption induced by iE-DAP. NOD1-specific shRNA also inhibited the activation of the NOD1/NF-κB signaling pathway and reversed the inflammatory response and TJ injury in iE-DAP-treated BMECs. Above results suggest that iE-DAP activated the NF-κB and MLCK signaling pathway in NOD1-dependent manner, which promoted the transcription of inflammatory cytokines and altered the expression and distribution of tight junction proteins, finally caused inflammatory response and TJ disruption. This study might provide theoretical basis and scientific support for the prevention and treatment of mastitis.
Collapse
|
31
|
Wang Y, Wang L, Meng M, Huo R, Ma N, Chang G, Shen X. High concentrate diet induced inflammatory response and tight junction disruption in the mammary gland of dairy cows. Microb Pathog 2023; 176:105996. [PMID: 36709006 DOI: 10.1016/j.micpath.2023.105996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Revised: 01/14/2023] [Accepted: 01/16/2023] [Indexed: 01/26/2023]
Abstract
This study aimed to investigate the effect and mechanism of a high concentrate (HC) diet on the inflammatory response and cellular tight junctions (TJs) in the mammary gland of dairy cows. Twelve lactating Holstein dairy cows were randomly assigned into low concentrate (LC) and HC groups (n = 6), which were fed with LC diet and HC diet respectively for 3 weeks. The HC diet lead to subacute ruminant acidosis with a rumen pH < 5.6 more than 3 h daily. The HC diet triggered an inflammatory response with increased levels of inflammatory cytokines in the lacteal vein, upregulated expression of inflammation-related genes, elevated activity of myeloperoxidase, and inflammatory cells infiltration in the mammary gland. Furthermore, the HC diet induced the activation of nuclear factor kappa B (NF-κB) and mitogen-activated protein kinase (MAPK) signaling pathways with enhanced phosphorylation ratios of NF-κB P65, inhibitor of NF-κB (IκB), P38 and extracellular signal-regulated kinase 1/2 (ERK1/2) as well as decreased ratios of DNA methylation and chromatin compaction of genes coding for proinflammatory cytokines, which contributed to the upregulation of proinflammatory cytokine expression. The HC diet also destroyed the integrity of TJ with discontinuous and decreased expression levels of zonula occludens-1, Occludin, Claudin-4 and increased expression level of Claudin-1 in the mammary epithelial cells compared with LC group. Conclusively, the HC diet induced the activation of NF-κB and MAPK signaling pathways and epigenetic modifications, promoted the transcription of proinflammatory cytokines, and finally caused inflammatory response and TJ disruption in the mammary gland of dairy cows.
Collapse
Affiliation(s)
- Yan Wang
- Ministry of Education Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, PR China
| | - Lairong Wang
- Ministry of Education Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, PR China
| | - Meijuan Meng
- Ministry of Education Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, PR China
| | - Ran Huo
- Ministry of Education Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, PR China
| | - Nana Ma
- Ministry of Education Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, PR China
| | - Guangjun Chang
- Ministry of Education Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, PR China
| | - Xiangzhen Shen
- Ministry of Education Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, PR China.
| |
Collapse
|
32
|
Novichkova E, Nayak S, Boussiba S, Gopas J, Zilberg D, Khozin-Goldberg I. Dietary Application of the Microalga Lobosphaera incisa P127 Reduces Severity of Intestinal Inflammation, Modulates Gut-Associated Gene Expression, and Microbiome in the Zebrafish Model of IBD. Mol Nutr Food Res 2023; 67:e2200253. [PMID: 36683256 DOI: 10.1002/mnfr.202200253] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 11/30/2022] [Indexed: 01/24/2023]
Abstract
SCOPE Microalgae are an emerging nutritional resource of biomolecules with potential to alleviate gut inflammation. The study explores the anti-inflammatory and immunomodulatory potential of the microalga Lobosphaera incisa P127, which accumulates a rare omega-6 LC-PUFA dihomo-ɣ-linolenic acid (DGLA) under nitrogen starvation. The therapeutic potential of dietary supplementation with P127 is investigated in the zebrafish model of IBD (TNBS-induced colitis). METHODS AND RESULTS Guts are sampled from zebrafish fed experimental diets for 4 weeks, before and 24 h after TNBS challenge. Diets containing 15% non-starved (Ns) and 7.5% and 15% N-starved (St) algal biomass significantly attenuate the severity of gut injury and goblet cell depletion. In contrast, diets containing 7.5% Ns and DGLA ethyl ester have no effect on gut condition. Fish fed 15% St, high-DGLA biomass, have the fewest individuals with pathological alterations in the gut. Dietary inclusion of Ns and St distinctly modulates gut-associated expression of the immune and inflammatory genes. Fish fed 15% Ns biomass display a coordinated boost in immune gene expression and show major changes in the gut microbiome prior challenge. CONCLUSION Dietary inclusion of L. incisa biomass at two physiological states, ameliorates TNBS-induced gut inflammation, suggesting the synergistic beneficial effects of biomass components not limited to DGLA.
Collapse
Affiliation(s)
- Ekaterina Novichkova
- The French Associates Institute for Agriculture and Biotechnology of Drylands, The Jacob Blaustein Institutes for Desert Research, Ben-Gurion University of the Negev, Sede Boqer Campus, Midreshet Ben-Gurion, 8499000, Israel
- The Albert Katz International School for Desert Studies, The Jacob Blaustein Institutes for Desert Research, Ben-Gurion University of the Negev, Sede Boqer Campus, Midreshet Ben-Gurion, 8499000, Israel
| | - Sagar Nayak
- The French Associates Institute for Agriculture and Biotechnology of Drylands, The Jacob Blaustein Institutes for Desert Research, Ben-Gurion University of the Negev, Sede Boqer Campus, Midreshet Ben-Gurion, 8499000, Israel
- The Jacob Blaustein Center for Scientific Cooperation, The Jacob Blaustein Institutes for Desert Research, Ben-Gurion University of the Negev, Sede Boqer Campus, Midreshet Ben-Gurion, 8499000, Israel
| | - Sammy Boussiba
- The French Associates Institute for Agriculture and Biotechnology of Drylands, The Jacob Blaustein Institutes for Desert Research, Ben-Gurion University of the Negev, Sede Boqer Campus, Midreshet Ben-Gurion, 8499000, Israel
| | - Jacob Gopas
- Department of Microbiology and Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, 8400501, Israel
| | - Dina Zilberg
- The French Associates Institute for Agriculture and Biotechnology of Drylands, The Jacob Blaustein Institutes for Desert Research, Ben-Gurion University of the Negev, Sede Boqer Campus, Midreshet Ben-Gurion, 8499000, Israel
| | - Inna Khozin-Goldberg
- The French Associates Institute for Agriculture and Biotechnology of Drylands, The Jacob Blaustein Institutes for Desert Research, Ben-Gurion University of the Negev, Sede Boqer Campus, Midreshet Ben-Gurion, 8499000, Israel
| |
Collapse
|
33
|
Liu J, Lu R, Zheng X, Hou W, Wu X, Zhao H, Wang G, Tian T. Establishment of a gut-on-a-chip device with controllable oxygen gradients to study the contribution of Bifidobacterium bifidum to inflammatory bowel disease. Biomater Sci 2023; 11:2504-2517. [PMID: 36779280 DOI: 10.1039/d2bm01490d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Supplemental Bifidobacterium has been shown to aid in the prevention, alleviation, and treatment of inflammatory bowel disease (IBD), but the progression and mechanisms are largely unstudied, partly because of a lack of appropriate models. In vitro human gut models must accurately recreate oxygen concentration gradients consistent with those in vivo to mimic gene expression, metabolism, and host-microbiome interactions. A non-equipment-intensive and inexpensive method for constructing the gut-on-a-chip with physiological oxygen concentration gradients remains challenging. Here, we propose a simple strategy using numerical simulations in a dual-channel gut-on-a-chip to guide chip design and achieve controllable oxygen gradients. By varying the size of microchannels, blocking the oxygen penetration of the polydimethylsiloxane layer at a given location, and controlling the flow of hypoxic/aerobic media, this strategy creates steep gradients across the intestinal epithelium. IBD symptoms were induced on the chip by tumor necrosis factor-α and lipopolysaccharide treatment. Bifidobacterium bifidum has been validated to contribute to the stability of the intestinal epithelial barrier, including preventing epithelial barrier disruption and promoting the repair of damaged intestinal epithelial cell monolayers. These effects may be associated with the co-localization of Bifidobacterium bifidum and ZO-1. This simple but robust approach for designing microfluidic devices is applicable to various organs-on-chips in which fluid dynamics and concentration profiles between different media must be considered. With the customized chip, the integration of activated Bifidobacterium bifidum provides an initial step toward developing a multi-factorial IBD platform. The approach could be scaled up for disease modeling, high-throughput drug screening and personalized medicine.
Collapse
Affiliation(s)
- Jun Liu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China.
| | - Ronghao Lu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China.
| | - Xiaolin Zheng
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China.
| | - Wensheng Hou
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China.
| | - Xiaoying Wu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China.
| | - Hezhao Zhao
- Department of Gastrointestinal Surgery, Chongqing University Cancer Hospital, Chongqing University, Chongqing, 400030, China
| | - Guixue Wang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China.
| | - Tian Tian
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China.
| |
Collapse
|
34
|
Dvořák Z, Li H, Mani S. Microbial Metabolites as Ligands to Xenobiotic Receptors: Chemical Mimicry as Potential Drugs of the Future. Drug Metab Dispos 2023; 51:219-227. [PMID: 36184080 PMCID: PMC9900867 DOI: 10.1124/dmd.122.000860] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 08/28/2022] [Accepted: 09/19/2022] [Indexed: 01/31/2023] Open
Abstract
Xenobiotic receptors, such as the pregnane X receptor, regulate multiple host physiologic pathways including xenobiotic metabolism, certain aspects of cellular metabolism, and innate immunity. These ligand-dependent nuclear factors regulate gene expression via genomic recognition of specific promoters and transcriptional activation of the gene. Natural or endogenous ligands are not commonly associated with this class of receptors; however, since these receptors are expressed in a cell-type specific manner in the liver and intestines, there has been significant recent effort to characterize microbially derived metabolites as ligands for these receptors. In general, these metabolites are thought to be weak micromolar affinity ligands. This journal anniversary minireview focuses on recent efforts to derive potentially nontoxic microbial metabolite chemical mimics that could one day be developed as drugs combating xenobiotic receptor-modifying pathophysiology. The review will include our perspective on the field and recommend certain directions for future research. SIGNIFICANCE STATEMENT: Xenobiotic receptors (XRs) regulate host drug metabolism, cellular metabolism, and immunity. Their presence in host intestines allows them to function not only as xenosensors but also as a response to the complex metabolic environment present in the intestines. Specifically, this review focuses on describing microbial metabolite-XR interactions and the translation of these findings toward discovery of novel chemical mimics as potential drugs of the future for diseases such as inflammatory bowel disease.
Collapse
Affiliation(s)
- Zdeněk Dvořák
- Department of Cell Biology and Genetics, Palacký University, Olomouc, Czech Republic (Z.D.); Departments of Medicine (H.L., S.M.), Molecular Pharmacology (S.M.), and Genetics (S.M.), Albert Einstein College of Medicine, Bronx, New York, USA
| | - Hao Li
- Department of Cell Biology and Genetics, Palacký University, Olomouc, Czech Republic (Z.D.); Departments of Medicine (H.L., S.M.), Molecular Pharmacology (S.M.), and Genetics (S.M.), Albert Einstein College of Medicine, Bronx, New York, USA
| | - Sridhar Mani
- Department of Cell Biology and Genetics, Palacký University, Olomouc, Czech Republic (Z.D.); Departments of Medicine (H.L., S.M.), Molecular Pharmacology (S.M.), and Genetics (S.M.), Albert Einstein College of Medicine, Bronx, New York, USA
| |
Collapse
|
35
|
Li H, Han R, Yong F, Fan Y, Zhao B, Hu X, Zhang T, Che D. The protective effect of Eleutheroside E against the mechanical barrier dysfunction triggered by lipopolysaccharide in IPEC-J2 cells. Res Vet Sci 2023; 154:1-7. [PMID: 36375269 DOI: 10.1016/j.rvsc.2022.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 10/26/2022] [Accepted: 11/01/2022] [Indexed: 11/06/2022]
Abstract
Eleutheroside E (EE) exhibits immunocompetence, antioxidant, and anti-inflammatory activity. Lipopolysaccharide (LPS) can elicit a strong immune response. In vitro experiments were used to explore whether EE protects intestinal porcine jejunum epithelial cells (IPEC-J2) barriers from LPS stress. The experiment was divided into group C (control group: complete medium), group E (group C + 0.1 mg/mL EE), group L (group C + 10 μg/mL LPS), and group EL (adding 0.1 mg/mL EE for 6 h, and then adding 10 μg/mL LPS for culture). Finally, the cell proliferation, permeability, mRNA expression of cytokines, mRNA and protein expression of tight junctions (TJs) were analyzed. The result show that, when compared to the C group, EE significantly promoted the proliferation of IPEC-J2 at 58 h and showed low permeability (P < 0.05), the anti-inflammatory cytokines IL-10 and TGF-β mRNA expression were increased extremely significantly, the inflammatory cytokines IL-6, TNF-α, and IFN-γ mRNA expression were extremely significantly decreased (P < 0.01), the mRNA and protein expression of TJ were significantly increased in group E (P < 0.05). However, LPS showed a damaging effect. EL group compared with L group, the cell index (CI) value was higher at 58 h (P < 0.05), the permeability was significantly lower (P < 0.05), the mRNA expressions of the inflammatory cytokines were down-regulated(P < 0.01), and the TJ mRNA and protein relative expression were increased (P < 0.05). In summary, the addition of EE protects the LPS-induced increase in permeability of IPEC-J2, potentially by expressing high levels of TJ proteins and inhibiting the increase of inflammatory cytokines.
Collapse
Affiliation(s)
- Huijuan Li
- College of Animal Science and Technology, Jilin Agricultural University, No. 2888 Xincheng Street, Changchun 130118, China; Ministry of Education Laboratory of Animal Production and Quality Security, Jilin Agricultural University, Changchun, China; Jilin Provincial Key Laboratory of Animal Nutrition and Feed Science, Jilin Agricultural University, Changchun, China
| | - Rui Han
- College of Animal Science and Technology, Jilin Agricultural University, No. 2888 Xincheng Street, Changchun 130118, China; Ministry of Education Laboratory of Animal Production and Quality Security, Jilin Agricultural University, Changchun, China; Jilin Provincial Key Laboratory of Animal Nutrition and Feed Science, Jilin Agricultural University, Changchun, China
| | - Feng Yong
- College of Animal Science and Technology, Jilin Agricultural University, No. 2888 Xincheng Street, Changchun 130118, China; Ministry of Education Laboratory of Animal Production and Quality Security, Jilin Agricultural University, Changchun, China; Jilin Provincial Key Laboratory of Animal Nutrition and Feed Science, Jilin Agricultural University, Changchun, China
| | - Yueli Fan
- College of Animal Science and Technology, Jilin Agricultural University, No. 2888 Xincheng Street, Changchun 130118, China; Ministry of Education Laboratory of Animal Production and Quality Security, Jilin Agricultural University, Changchun, China; Jilin Provincial Key Laboratory of Animal Nutrition and Feed Science, Jilin Agricultural University, Changchun, China
| | - Bao Zhao
- College of Animal Science and Technology, Jilin Agricultural University, No. 2888 Xincheng Street, Changchun 130118, China; Ministry of Education Laboratory of Animal Production and Quality Security, Jilin Agricultural University, Changchun, China; Jilin Provincial Key Laboratory of Animal Nutrition and Feed Science, Jilin Agricultural University, Changchun, China
| | - Xiaocai Hu
- College of Animal Science and Technology, Jilin Agricultural University, No. 2888 Xincheng Street, Changchun 130118, China; Ministry of Education Laboratory of Animal Production and Quality Security, Jilin Agricultural University, Changchun, China; Jilin Provincial Key Laboratory of Animal Nutrition and Feed Science, Jilin Agricultural University, Changchun, China
| | - Tianrui Zhang
- College of Animal Science and Technology, Jilin Agricultural University, No. 2888 Xincheng Street, Changchun 130118, China; Ministry of Education Laboratory of Animal Production and Quality Security, Jilin Agricultural University, Changchun, China; Jilin Provincial Key Laboratory of Animal Nutrition and Feed Science, Jilin Agricultural University, Changchun, China.
| | - Dongsheng Che
- College of Animal Science and Technology, Jilin Agricultural University, No. 2888 Xincheng Street, Changchun 130118, China; Ministry of Education Laboratory of Animal Production and Quality Security, Jilin Agricultural University, Changchun, China; Jilin Provincial Key Laboratory of Animal Nutrition and Feed Science, Jilin Agricultural University, Changchun, China.
| |
Collapse
|
36
|
Jiang S, Miao Z. High-fat diet induces intestinal mucosal barrier dysfunction in ulcerative colitis: emerging mechanisms and dietary intervention perspective. Am J Transl Res 2023; 15:653-677. [PMID: 36915785 PMCID: PMC10006746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Accepted: 01/09/2023] [Indexed: 03/16/2023]
Abstract
The incidence of ulcerative colitis (UC) is increasing worldwide, but its pathogenesis remains largely unclear. The intestinal mucosa is a barrier that maintains the stability of the body's internal environment, and dysfunction of this barrier leads to the occurrence and aggravation of UC. A high-fat diet (HFD) contains more animal fat and low fiber, and accumulating evidence has shown that long-term intake of an HFD is associated with UC. The mechanism linking an HFD with intestinal mucosal barrier disruption is multifactorial, and it typically involves microbiota dysbiosis and altered metabolism of fatty acids, bile acids, and tryptophan. Dysbiosis-induced metabolic changes can enhance intestinal permeability through multiple pathways. These changes modulate the programmed death of intestinal epithelial cells, inhibit the secretion of goblet cells and Paneth cells, and impair intercellular interactions. Gut metabolites can also induce intestinal immune imbalance by stimulating multiple proinflammatory signaling pathways and decreasing the effect of anti-inflammatory immune cells. In this review, we critically analyze the molecular mechanisms by which an HFD disrupts the intestinal mucosal barrier (IMB) and contributes to the development of UC. We also discuss the application and future direction of dietary intervention in the treatment of the IMB and prevention of UC.
Collapse
Affiliation(s)
- Shijing Jiang
- First Clinical Medical College, Nanjing University of Chinese Medicine Nanjing, Jiangsu, China
| | - Zhiwei Miao
- Department of Gastroenterology, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine Zhangjiagang, Suzhou, Jiangsu, China
| |
Collapse
|
37
|
Mavrogeni ME, Asadpoor M, Henricks PAJ, Keshavarzian A, Folkerts G, Braber S. Direct Action of Non-Digestible Oligosaccharides against a Leaky Gut. Nutrients 2022; 14:4699. [PMID: 36364961 PMCID: PMC9655944 DOI: 10.3390/nu14214699] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 11/01/2022] [Accepted: 11/02/2022] [Indexed: 10/28/2023] Open
Abstract
The epithelial monolayer is the primary determinant of mucosal barrier function, and tight junction (TJ) complexes seal the paracellular space between the adjacent epithelial cells and represent the main "gate-keepers" of the paracellular route. Impaired TJ functionality results in increased permeation of the "pro-inflammatory" luminal contents to the circulation that induces local and systemic inflammatory and immune responses, ultimately triggering and/or perpetuating (chronic) systemic inflammatory disorders. Increased gut leakiness is associated with intestinal and systemic disease states such as inflammatory bowel disease and neurodegenerative diseases such as Parkinson's disease. Modulation of TJ dynamics is an appealing strategy aiming at inflammatory conditions associated with compromised intestinal epithelial function. Recently there has been a growing interest in nutraceuticals, particularly in non-digestible oligosaccharides (NDOs). NDOs confer innumerable health benefits via microbiome-shaping and gut microbiota-related immune responses, including enhancement of epithelial barrier integrity. Emerging evidence supports that NDOs also exert health-beneficial effects on microbiota independently via direct interactions with intestinal epithelial and immune cells. Among these valuable features, NDOs promote barrier function by directly regulating TJs via AMPK-, PKC-, MAPK-, and TLR-associated pathways. This review provides a comprehensive overview of the epithelial barrier-protective effects of different NDOs with a special focus on their microbiota-independent modulation of TJs.
Collapse
Affiliation(s)
- Maria Eleni Mavrogeni
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584 CG Utrecht, The Netherlands
| | - Mostafa Asadpoor
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584 CG Utrecht, The Netherlands
| | - Paul A. J. Henricks
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584 CG Utrecht, The Netherlands
| | - Ali Keshavarzian
- Division of Gastroenterology, Department of Internal Medicine, Rush University Medical Center, Chicago, IL 60612, USA
| | - Gert Folkerts
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584 CG Utrecht, The Netherlands
| | - Saskia Braber
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584 CG Utrecht, The Netherlands
| |
Collapse
|
38
|
Lonardo A, Mantovani A, Petta S, Carraro A, Byrne CD, Targher G. Metabolic mechanisms for and treatment of NAFLD or NASH occurring after liver transplantation. Nat Rev Endocrinol 2022; 18:638-650. [PMID: 35840803 DOI: 10.1038/s41574-022-00711-5] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/07/2022] [Indexed: 11/08/2022]
Abstract
The rising tide of non-alcoholic fatty liver disease (NAFLD) associated with the obesity epidemic is a major health concern worldwide. NAFLD - specifically its more advanced form, non-alcoholic steatohepatitis (NASH)-related cirrhosis - is now the fastest growing indication for liver transplantation in the USA and Europe. Although the short-term and mid-term overall survival rates of patients who receive a liver transplant for NASH-related cirrhosis are essentially similar to those of patients who receive a transplant for other liver indications, recipients with NASH-related cirrhosis have an increased risk of waiting-list mortality and of developing recurrent liver disease and cardiometabolic complications in the longer term after liver transplantation. This Review provides a brief overview of the epidemiology of NAFLD and NASH and the occurrence of NAFLD or NASH in patients after liver transplantation for NASH and other liver indications. It also discusses the putative metabolic mechanisms underlying the emergence of NAFLD or NASH after liver transplantation as well as optimal therapeutic approaches for recipients of liver transplants, including the management of cardiometabolic comorbidities, tailored immunosuppression, lifestyle changes and pharmacotherapy for NAFLD.
Collapse
Affiliation(s)
- Amedeo Lonardo
- Metabolic Syndrome Unit, University of Modena, Modena, Italy
| | - Alessandro Mantovani
- Section of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Verona, Verona, Italy
| | - Salvatore Petta
- Section of Gastroenterology and Hepatology, PROMISE, University of Palermo, Palermo, Italy
| | - Amedeo Carraro
- Liver Transplant Unit, University of Verona, Verona, Italy
| | - Christopher D Byrne
- Nutrition and Metabolism, Faculty of Medicine, University of Southampton, Southampton, UK
- Southampton National Institute for Health Research Biomedical Research Centre, University Hospital Southampton, Southampton General Hospital, Southampton, UK
| | - Giovanni Targher
- Section of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Verona, Verona, Italy.
| |
Collapse
|
39
|
Effects of Butyrate Supplementation on Inflammation and Kidney Parameters in Type 1 Diabetes: A Randomized, Double-Blind, Placebo-Controlled Trial. J Clin Med 2022; 11:jcm11133573. [PMID: 35806857 PMCID: PMC9267418 DOI: 10.3390/jcm11133573] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 04/27/2022] [Accepted: 06/09/2022] [Indexed: 12/19/2022] Open
Abstract
Type 1 diabetes is associated with increased intestinal inflammation and decreased abundance of butyrate-producing bacteria. We investigated the effect of butyrate on inflammation, kidney parameters, HbA1c, serum metabolites and gastrointestinal symptoms in persons with type 1 diabetes, albuminuria and intestinal inflammation. We conducted a randomized placebo-controlled, double-blind, parallel clinical study involving 53 participants randomized to 3.6 g sodium butyrate daily or placebo for 12 weeks. The primary endpoint was the change in fecal calprotectin. Additional endpoints were the change in fecal short chain fatty acids, intestinal alkaline phosphatase activity and immunoglobulins, serum lipopolysaccharide, CRP, albuminuria, kidney function, HbA1c, metabolites and gastrointestinal symptoms. The mean age was 54 ± 13 years, and the median [Q1:Q3] urinary albumin excretion was 46 [14:121] mg/g. The median fecal calprotectin in the butyrate group was 48 [26:100] μg/g at baseline, and the change was −1.0 [−20:10] μg/g; the median in the placebo group was 61 [25:139] μg/g at baseline, and the change was −12 [−95:1] μg/g. The difference between the groups was not significant (p = 0.24); neither did we find an effect of butyrate compared to placebo on the other inflammatory markers, kidney parameters, HbA1c, metabolites nor gastrointestinal symptoms. Twelve weeks of butyrate supplementation did not reduce intestinal inflammation in persons with type 1 diabetes, albuminuria and intestinal inflammation.
Collapse
|
40
|
Zhao H, Chen R, Zheng D, Xiong F, Jia F, Liu J, Zhang L, Zhang N, Zhu S, Liu Y, Zhao L, Liu X. Modified Banxia Xiexin Decoction Ameliorates Polycystic Ovarian Syndrome With Insulin Resistance by Regulating Intestinal Microbiota. Front Cell Infect Microbiol 2022; 12:854796. [PMID: 35619648 PMCID: PMC9127304 DOI: 10.3389/fcimb.2022.854796] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 04/11/2022] [Indexed: 12/12/2022] Open
Abstract
Objective To analyze the characteristics of the intestinal microbiota of polycystic ovarian syndrome with insulin resistance (PCOS-IR) and explore the possible mechanism of modified Banxia Xiexin Decoction in the treatment of PCOS-IR. Methods A total of 17 specific pathogen-free (SPF) female Sprague–Dawley (SD) rats, aged 21 days, were selected and randomly divided into the control group (group Z, n = 6), model group (group M, n = 6), and treatment group (group A, n = 5). Letrozole combined with a high-fat diet was used to induce the PCOS-IR model. Rats in group A were treated with modified Banxia Xiexin Decoction for 2 weeks after the end of modeling; then the characteristics of reproductive, metabolic, inflammatory, and intestinal microbiota were compared among three groups. Results The PCOS-IR model had an imbalance of intestinal microbiota, and the enriched microbiota was mainly class Coriobacteria, order Clostridiales, and genus Clostridium_sensu_stricto_1. Modified Banxia Xiexin Decoction can regulate the disorder of intestinal microbiota diversity, significantly increase the abundance of phyla Verrucomicrobiota Proteobacteria and genera Akkermansia and Blautia, and decrease the abundance of genus Clostridium_sensu_stricto_1. Conclusion Genus Clostridium_sensu_stricto_1 might be the pivotal pathogenic bacteria of PCOS-IR. Modified Banxia Xiexin Decoction may ameliorate PCOS-IR by regulating intestinal microbiota imbalance and improving metabolic disorders.
Collapse
Affiliation(s)
- Hongyu Zhao
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Rufeng Chen
- Graduate College, Beijing University of Chinese Medicine, Beijing, China
| | - Dongxue Zheng
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Feng Xiong
- Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing, China
| | - Fan Jia
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jinyuan Liu
- Graduate College, Beijing University of Chinese Medicine, Beijing, China
| | - Lili Zhang
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Nana Zhang
- Graduate College, Beijing University of Chinese Medicine, Beijing, China
| | - Shiqin Zhu
- Graduate College, Beijing University of Chinese Medicine, Beijing, China
| | - Yongmei Liu
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Linhua Zhao
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xinmin Liu
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- *Correspondence: Xinmin Liu,
| |
Collapse
|
41
|
Li Y, Liu N, Ge Y, Yang Y, Ren F, Wu Z. Tryptophan and the innate intestinal immunity: Crosstalk between metabolites, host innate immune cells and microbiota. Eur J Immunol 2022; 52:856-868. [PMID: 35362153 DOI: 10.1002/eji.202149401] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Revised: 12/17/2021] [Accepted: 01/20/2022] [Indexed: 11/11/2022]
Abstract
The intestinal mucosal barrier is critical for the absorption of nutrients and the health of both humans and animals. Recent publications from clinical and experimental studies have shown the importanceof the nutrients-bacteria-host interaction for the intestinal homeostasis. Dysfunction of these interactions has been reported to be associated with metabolic disorders and development of intestinal diseases, such as the irritable bowel syndrome and inflammatory bowel diseases. Tryptophan and its metabolites, including kynurenine, kynurenic acid, and 5-hydroxytrptamine, can influence the proliferation of enterocytes, intestinal integrity and immune response, as well as intestinal microbiota, therefore regulating and contributing to the intestinal health. In this review, we highlight recent findings on the effect of tryptophan and its metabolites on the mucosal barrier and intestinal homeostasis and its regulation of innate immune response. Moreover, we present the signaling pathways related to Trp metabolism, such as mammalian target of rapamycin, aryl hydrocarbon receptor, and pregnane X receptor, that contribute to the intestinal homeostasis and discuss future perspectives on spontaneous interference in host tryptophan metabolism as potential clinical strategies of intestinal diseases. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Yunke Li
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing, 100193, China
| | - Ning Liu
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing, 100193, China
| | - Yao Ge
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing, 100193, China
| | - Ying Yang
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, 100193, China
| | - Fazheng Ren
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing, 100193, China
| | - Zhenlong Wu
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing, 100193, China.,State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, 100193, China
| |
Collapse
|
42
|
Cordycepin ameliorates acute hypobaric hypoxia induced blood-brain barrier disruption, and cognitive impairment partly by suppressing the TLR4/NF-κB/MMP-9 pathway in the adult rats. Eur J Pharmacol 2022; 924:174952. [DOI: 10.1016/j.ejphar.2022.174952] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 04/04/2022] [Accepted: 04/06/2022] [Indexed: 02/07/2023]
|
43
|
Mohr AE, Crawford M, Jasbi P, Fessler S, Sweazea KL. Lipopolysaccharide and the gut microbiota: Considering structural variation. FEBS Lett 2022; 596:849-875. [PMID: 35262962 DOI: 10.1002/1873-3468.14328] [Citation(s) in RCA: 67] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 02/16/2022] [Accepted: 02/17/2022] [Indexed: 11/10/2022]
Abstract
Systemic inflammation is associated with chronic disease and is purported to be a main pathogenic mechanism underlying metabolic conditions. Microbes harbored in the host gastrointestinal tract release signaling byproducts from their cell wall, such as lipopolysaccharides (LPS), which can act locally and, after crossing the gut barrier and entering circulation, also systemically. Defined as metabolic endotoxemia, elevated concentrations of LPS in circulation are associated with metabolic conditions and chronic disease. As such, measurement of LPS is highly prevalent in animal and human research investigating these states. Indeed, LPS can be a potent stimulant of host immunity but this response depends on the microbial species' origin, a parameter often overlooked in both preclinical and clinical investigations. Indeed, the lipid A portion of LPS is mutable and comprises the main virulence and endotoxic component, thus contributing to the structural and functional diversity among LPSs from microbial species. In this review, we discuss how such structural differences in LPS can induce differential immunological responses in the host.
Collapse
Affiliation(s)
- Alex E Mohr
- College of Health Solutions, Arizona State University, Phoenix, Arizona, United States of America
| | - Meli'sa Crawford
- Biomedical Sciences, University of Riverside, California, Riverside, California, United States of America
| | - Paniz Jasbi
- College of Health Solutions, Arizona State University, Phoenix, Arizona, United States of America
| | - Samantha Fessler
- College of Health Solutions, Arizona State University, Phoenix, Arizona, United States of America
| | - Karen L Sweazea
- College of Health Solutions, Arizona State University, Phoenix, Arizona, United States of America.,School of Life Sciences, Arizona State University, Tempe, Arizona, United States of America
| |
Collapse
|
44
|
Implication of Intestinal Barrier Dysfunction in Gut Dysbiosis and Diseases. Biomedicines 2022; 10:biomedicines10020289. [PMID: 35203499 PMCID: PMC8869546 DOI: 10.3390/biomedicines10020289] [Citation(s) in RCA: 154] [Impact Index Per Article: 51.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 01/21/2022] [Accepted: 01/24/2022] [Indexed: 02/04/2023] Open
Abstract
The intestinal mucosal barrier, also referred to as intestinal barrier, is widely recognized as a critical player in gut homeostasis maintenance as it ensures the complex crosstalk between gut microbes (both commensals and pathogens) and the host immune system. Highly specialized epithelial cells constantly cope with several protective and harmful agents to maintain the multiple physiological functions of the barrier as well as its integrity. However, both genetic defects and environmental factors can break such equilibrium, thus promoting gut dysbiosis, dysregulated immune-inflammatory responses, and even the development of chronic pathological conditions. Here, we review and discuss the molecular and cellular pathways underlying intestinal barrier structural and functional homeostasis, focusing on potential alterations that may undermine this fine balance.
Collapse
|
45
|
The Role of Gut-Derived Lipopolysaccharides and the Intestinal Barrier in Fatty Liver Diseases. J Gastrointest Surg 2022; 26:671-683. [PMID: 34734369 PMCID: PMC8926958 DOI: 10.1007/s11605-021-05188-7] [Citation(s) in RCA: 109] [Impact Index Per Article: 36.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Accepted: 10/14/2021] [Indexed: 02/06/2023]
Abstract
BACKGROUND Hepatosteatosis is the earliest stage in the pathogenesis of nonalcoholic fatty (NAFLD) and alcoholic liver disease (ALD). As NAFLD is affecting 10-24% of the general population and approximately 70% of obese patients, it carries a large economic burden and is becoming a major reason for liver transplantation worldwide. ALD is a major cause of morbidity and mortality causing 50% of liver cirrhosis and 10% of liver cancer related death. Increasing evidence has accumulated that gut-derived factors play a crucial role in the development and progression of chronic liver diseases. METHODS A selective literature search was conducted in Medline and PubMed, using the terms "nonalcoholic fatty liver disease," "alcoholic liver disease," "lipopolysaccharide," "gut barrier," and "microbiome." RESULTS Gut dysbiosis and gut barrier dysfunction both contribute to chronic liver disease by abnormal regulation of the gut-liver axis. Thereby, gut-derived lipopolysaccharides (LPS) are a key factor in inducing the inflammatory response of liver tissue. The review further underlines that endotoxemia is observed in both NAFLD and ALD patients. LPS plays an important role in conducting liver damage through the LPS-TLR4 signaling pathway. Treatments targeting the gut microbiome and the gut barrier such as fecal microbiota transplantation (FMT), probiotics, prebiotics, synbiotics, and intestinal alkaline phosphatase (IAP) represent potential treatment modalities for NAFLD and ALD. CONCLUSIONS The gut-liver axis plays an important role in the development of liver disease. Treatments targeting the gut microbiome and the gut barrier have shown beneficial effects in attenuating liver inflammation and need to be further investigated.
Collapse
|
46
|
Lad N, Murphy A, Parenti C, Nelson C, Williams N, Sharpe G, McTernan P. Asthma and obesity: endotoxin another insult to add to injury? Clin Sci (Lond) 2021; 135:2729-2748. [PMID: 34918742 PMCID: PMC8689194 DOI: 10.1042/cs20210790] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 11/29/2021] [Accepted: 12/06/2021] [Indexed: 12/20/2022]
Abstract
Low-grade inflammation is often an underlying cause of several chronic diseases such as asthma, obesity, cardiovascular disease, and type 2 diabetes mellitus (T2DM). Defining the mediators of such chronic low-grade inflammation often appears dependent on which disease is being investigated. However, downstream systemic inflammatory cytokine responses in these diseases often overlap, noting there is no doubt more than one factor at play to heighten the inflammatory response. Furthermore, it is increasingly believed that diet and an altered gut microbiota may play an important role in the pathology of such diverse diseases. More specifically, the inflammatory mediator endotoxin, which is a complex lipopolysaccharide (LPS) derived from the outer membrane cell wall of Gram-negative bacteria and is abundant within the gut microbiota, and may play a direct role alongside inhaled allergens in eliciting an inflammatory response in asthma. Endotoxin has immunogenic effects and is sufficiently microscopic to traverse the gut mucosa and enter the systemic circulation to act as a mediator of chronic low-grade inflammation in disease. Whilst the role of endotoxin has been considered in conditions of obesity, cardiovascular disease and T2DM, endotoxin as an inflammatory trigger in asthma is less well understood. This review has sought to examine the current evidence for the role of endotoxin in asthma, and whether the gut microbiota could be a dietary target to improve disease management. This may expand our understanding of endotoxin as a mediator of further low-grade inflammatory diseases, and how endotoxin may represent yet another insult to add to injury.
Collapse
Affiliation(s)
- Nikita Lad
- Department of Biosciences, School of Science and Technology, Nottingham Trent University, Nottingham, NG11 8NS, U.K
| | - Alice M. Murphy
- Department of Biosciences, School of Science and Technology, Nottingham Trent University, Nottingham, NG11 8NS, U.K
| | - Cristina Parenti
- SHAPE Research Centre, School of Science and Technology, Nottingham Trent University, Nottingham, NG11 8NS, U.K
| | - Carl P. Nelson
- Department of Biosciences, School of Science and Technology, Nottingham Trent University, Nottingham, NG11 8NS, U.K
| | - Neil C. Williams
- SHAPE Research Centre, School of Science and Technology, Nottingham Trent University, Nottingham, NG11 8NS, U.K
| | - Graham R. Sharpe
- SHAPE Research Centre, School of Science and Technology, Nottingham Trent University, Nottingham, NG11 8NS, U.K
| | - Philip G. McTernan
- Department of Biosciences, School of Science and Technology, Nottingham Trent University, Nottingham, NG11 8NS, U.K
| |
Collapse
|
47
|
Xiao L, Zhang WH, Huang Y, Huang P. Intestinal ischemia‑reperfusion induces the release of IL‑17A to regulate cell inflammation, apoptosis and barrier damage. Exp Ther Med 2021; 23:158. [PMID: 35069839 PMCID: PMC8753980 DOI: 10.3892/etm.2021.11081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 11/16/2021] [Indexed: 12/01/2022] Open
Abstract
Intestinal ischemia-reperfusion (I/R) injury promotes the release of IL-17A, and previous studies have indicated that TGF-β activated kinase 1 (TAK1) is an important signaling molecule in the regulatory function of IL-17A. The present study aimed to explore the potential effects of IL-17A release in intestinal I/R injury, and to investigate the underlying regulatory mechanisms. Initially, the expression levels of TAK1 and JNK in a hypoxia/reoxygenation model were determined, and the effects of TAK1-knockdown on JNK phosphorylation and the viability, inflammation, apoptosis and barrier function of Caco-2 cells were assessed using Cell Counting Kit-8, reverse transcription-quantitative PCR, TUNEL and transepithelial electrical resistance assays, respectively. Subsequently, an antibody targeting IL-17A was used, and the effects of the IL-17A antibody on the expression levels of TAK1 as well as cell viability, inflammation, apoptosis and barrier function were determined. The results of the present study demonstrated that TAK1-knockdown markedly reduced JNK phosphorylation and improved the levels of cell viability, inflammation, apoptosis and barrier function via the MAPK signaling pathway. In addition, treatment with the IL-17A antibody inhibited the expression of TAK1, and reversed the aforementioned effects of TAK1 on Caco-2 cells. In conclusion, intestinal I/R induces the release of IL-17A to regulate cell viability, inflammation, apoptosis and barrier damage via the TAK1/MAPK signaling pathway.
Collapse
Affiliation(s)
- Li Xiao
- Department of Pediatrics, South Medical University Affiliated Maternal and Child Health Hospital of Foshan, Foshan, Guangdong 528000, P.R. China
| | - Wan-Hua Zhang
- Department of Pediatrics, South Medical University Affiliated Maternal and Child Health Hospital of Foshan, Foshan, Guangdong 528000, P.R. China
| | - Yin Huang
- Department of Pediatrics, South Medical University Affiliated Maternal and Child Health Hospital of Foshan, Foshan, Guangdong 528000, P.R. China
| | - Peng Huang
- Department of Pediatrics, South Medical University Affiliated Maternal and Child Health Hospital of Foshan, Foshan, Guangdong 528000, P.R. China
| |
Collapse
|
48
|
Sun JN, Yu XY, Hou B, Ai M, Qi MT, Ma XY, Cai MJ, Gao M, Cai WW, Ni LL, Xu F, Zhou YT, Qiu LY. Vaccarin enhances intestinal barrier function in type 2 diabetic mice. Eur J Pharmacol 2021; 908:174375. [PMID: 34303666 DOI: 10.1016/j.ejphar.2021.174375] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 07/16/2021] [Accepted: 07/21/2021] [Indexed: 01/12/2023]
Abstract
AIMS Hyperglycemia and insulin resistance drive intestinal barrier dysfunction in type 2 diabetes (T2DM). Vaccarin, the main active component in the semen of traditional Chinese medicine Vaccaria has a definite effect on T2DM mice. The purpose of this study was to investigate whether vaccarin can enhance the intestinal barrier function in T2DM. MAIN METHODS The T2DM mice model was established by streptozocin and high-fat diet. Vaccarin at a dose of 1 mg/kg/day was administered. We evaluated the effects of vaccarin on gut microbiota and intestinal barrier function by 16S rRNA sequencing, Western blot, quantitative fluorescent PCR (qPCR), and morphological observation. Moreover, we constructed a single layer of the human intestinal epithelium model to determine the effect of vaccarin in vitro. RESULTS The experimental results showed that vaccarin alleviated inflammatory mediators in serum and intestinal tissue of mice (P < 0.05), which may depend on the improvement of tight junctions and gut microbiota (P < 0.05). Activation of extracellular regulated protein kinases (Erk1/2) stimulated myosin light chain kinase (MLCK). By inhibiting ERK expression (P < 0.05), vaccarin had similar effects to ERK inhibitors. In addition, the regulation of tight junction barriers also involved the abovementioned pathways in vivo. CONCLUSION Vaccarin could protect the intestinal barrier by inhibiting the ERK/MLCK signaling pathway and modulate the composition of the microbiota. These results suggested that vaccarin may be an effective candidate for improving intestinal barrier changes in T2DM.
Collapse
Affiliation(s)
- Jiang-Nan Sun
- Wuxi Medical School, Jiangnan University, Wuxi, 214122, Jiangsu Province, PR China
| | - Xiao-Yi Yu
- Wuxi Medical School, Jiangnan University, Wuxi, 214122, Jiangsu Province, PR China
| | - Bao Hou
- Wuxi Medical School, Jiangnan University, Wuxi, 214122, Jiangsu Province, PR China
| | - Min Ai
- Wuxi Medical School, Jiangnan University, Wuxi, 214122, Jiangsu Province, PR China
| | - Meng-Ting Qi
- School of Pharmaceutical Sciences, Jiangnan University, Wuxi, 214122, Jiangsu Province, PR China
| | - Xin-Yu Ma
- Wuxi Medical School, Jiangnan University, Wuxi, 214122, Jiangsu Province, PR China
| | - Ming-Jie Cai
- Wuxi Medical School, Jiangnan University, Wuxi, 214122, Jiangsu Province, PR China
| | - Min Gao
- Wuxi Medical School, Jiangnan University, Wuxi, 214122, Jiangsu Province, PR China
| | - Wei-Wei Cai
- Wuxi Medical School, Jiangnan University, Wuxi, 214122, Jiangsu Province, PR China
| | - Lu-Lu Ni
- Wuxi Medical School, Jiangnan University, Wuxi, 214122, Jiangsu Province, PR China
| | - Fei Xu
- Wuxi Medical School, Jiangnan University, Wuxi, 214122, Jiangsu Province, PR China
| | - Yue-Tao Zhou
- Wuxi Medical School, Jiangnan University, Wuxi, 214122, Jiangsu Province, PR China
| | - Li-Ying Qiu
- Wuxi Medical School, Jiangnan University, Wuxi, 214122, Jiangsu Province, PR China.
| |
Collapse
|
49
|
Li X, Li C, Li Y, Liu C, Liang X, Liu T, Liu Z. Sodium nitroprusside protects HFD induced gut dysfunction via activating AMPKα/SIRT1 signaling. BMC Gastroenterol 2021; 21:359. [PMID: 34600475 PMCID: PMC8487517 DOI: 10.1186/s12876-021-01934-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 09/14/2021] [Indexed: 12/14/2022] Open
Abstract
Background Activation of Adenosine 5′-monophosphate-activated protein kinase/Sirtuin1 (AMPK/SIRT1) exerts an effect in alleviating obesity and gut damage. Sodium nitroprusside (SNP), a nitric oxide (NO) donor, has been reported to activate AMPK. This study was to investigate the effect of SNP on HFD induced gut dysfunction and the mechanism. Methods SNP was applied on lipopolysaccharide (LPS) stimulated Caco-2 cell monolayers which mimicked intestinal epithelial barrier dysfunction and HFD-fed mice which were complicated by gut dysfunction. Then AMPKα/SIRT1 pathway and gut barrier indicators were investigated. Results SNP rescued the loss of tight junction proteins ZO-1 and occludin, the inhibition of AMPKα/SIRT1 in LPS stimulated Caco-2 cell monolayers, and the effects were not shown when AMPKa1 was knocked-down by siRNA. SNP also alleviated HFD induced obesity and gut dysfunction in mice, as indicated by the decreasing of intestinal permeability, the increasing expression of ZO-1 and occludin, the decreasing levels of pro-inflammatory cytokine IL-6, and the repairing of gut microbiota dysbiosis. These effects were complicated by the increased colonic NO content and the activated AMPKα/SIRT1 signaling. Conclusions The results may imply that SNP, as a NO donor, alleviates HFD induced gut dysfunction probably by activating the AMPKα/SIRT1 signaling pathway.
Collapse
Affiliation(s)
- Xiaomei Li
- Guangzhou Key Laboratory of Enhanced Recovery after Abdominal Surgery, Innovation Center for Advanced Interdisciplinary Medicine, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510700, China
| | - Chen Li
- Guangzhou Key Laboratory of Enhanced Recovery after Abdominal Surgery, Innovation Center for Advanced Interdisciplinary Medicine, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510700, China
| | - Yuanqi Li
- Guangzhou Key Laboratory of Enhanced Recovery after Abdominal Surgery, Innovation Center for Advanced Interdisciplinary Medicine, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510700, China
| | - Cong Liu
- Guangzhou Key Laboratory of Enhanced Recovery after Abdominal Surgery, Innovation Center for Advanced Interdisciplinary Medicine, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510700, China
| | - Xue Liang
- Guangzhou Key Laboratory of Enhanced Recovery after Abdominal Surgery, Innovation Center for Advanced Interdisciplinary Medicine, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510700, China.
| | - Ting Liu
- Guangzhou Key Laboratory of Enhanced Recovery after Abdominal Surgery, Innovation Center for Advanced Interdisciplinary Medicine, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510700, China.
| | - Zhihua Liu
- Guangzhou Key Laboratory of Enhanced Recovery after Abdominal Surgery, Innovation Center for Advanced Interdisciplinary Medicine, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510700, China.
| |
Collapse
|
50
|
Miller MR, Koch SR, Choi H, Lamb FS, Stark RJ. Apoptosis signal-regulating kinase 1 (ASK1) inhibition reduces endothelial cytokine production without improving permeability after toll-like receptor 4 (TLR4) challenge. Transl Res 2021; 235:115-128. [PMID: 33857660 PMCID: PMC8328918 DOI: 10.1016/j.trsl.2021.04.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 04/05/2021] [Accepted: 04/08/2021] [Indexed: 12/12/2022]
Abstract
Sepsis represents a life-threatening event often mediated by the host's response to pathogens such as gram-negative organisms, which release the proinflammatory lipopolysaccharide (LPS). Within the endothelium, the mitogen-activated protein kinase (MAPK) pathway is an important driver of endothelial injury during sepsis, of which oxidant-sensitive apoptosis signal-regulating kinase 1 (ASK1) is postulated to be a critical upstream regulator. We hypothesized that ASK1 would play a key role in endothelial inflammation during bacterial challenge. Utilizing RNA sequencing data from patients and cultured human microvascular endothelial cells (HMVECs), ASK1 expression was increased in sepsis and after LPS challenge. Two ASK1 inhibitors, GS444217 and MSC2023964A, reduced cytokine production in HMVECs following LPS stimulation, but had no effect on permeability as measured by transendothelial electrical resistance and intercellular space. MAPKs are known to interact with endothelial nitric oxide synthase (eNOS) and ASK1 expression levels correlated with eNOS expression in patients with septic shock. In addition, eNOS physically interacted with ASK1, though this interaction was not altered by ASK1 inhibition, nor did inhibition alter MAPK p38 activity. Instead, among MAPKs, ASK1 inhibition only impaired LPS-induced JNK phosphorylation. The reduction in JNK activation caused by ASK1 inhibition impaired JNK-mediated cytokine production without affecting permeability. Thus, LPS triggers JNK-dependent cytokine production that requires ASK1 activation, but both its effects on permeability and activation of p38 are ASK1-independent. These data demonstrate how distinct MAPK signaling pathways regulate endothelial inflammatory outputs during acute infectious challenge.
Collapse
Affiliation(s)
- Michael R Miller
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Stephen R Koch
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Hyehun Choi
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Fred S Lamb
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Ryan J Stark
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee.
| |
Collapse
|