1
|
Hesampour F, Bernstein CN, Ghia JE. Investigating the effect of neuro-immune communication on immune responses in health and disease: Exploring immunological disorders. Cell Immunol 2025; 413:104963. [PMID: 40378510 DOI: 10.1016/j.cellimm.2025.104963] [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: 01/30/2025] [Revised: 05/02/2025] [Accepted: 05/06/2025] [Indexed: 05/19/2025]
Abstract
Recent recognition of the intricate nervous-immune system interplay has prompted research into the specific cellular components involved in these interactions. Emerging evidence suggests that immune and neural cells collaborate within distinct units and act in concert to regulate tissue function and provide protection. These specialized neuro-immune cell units have been identified in diverse body tissues, ranging from lymphoid organs to the bone marrow and mucosal barriers. Their significance has become increasingly apparent as they are recognized as pivotal regulators influencing a broad spectrum of physiological and pathological processes. This recognition extends to critical roles in hematopoiesis, organ function, inflammatory responses, and intricate tissue repair processes. This review explores the bidirectional communication between the nervous and immune systems. The focus is on understanding the profound impact of this communication on immune cells within key anatomical sites, such as the bone marrow, gastrointestinal tract, and lymphoid organs. By examining these interactions, this review aims to shed light on how this intricate network operates under normal and pathological conditions, offering insights into the mechanisms underlying health and disease.
Collapse
Affiliation(s)
| | - Charles N Bernstein
- Internal Medicine, University of Manitoba, Winnipeg, Canada; Inflammatory Bowel Disease Clinical & Research Centre, University of Manitoba, Winnipeg, Canada
| | - Jean-Eric Ghia
- Immunology, University of Manitoba, Winnipeg, Canada; Internal Medicine, University of Manitoba, Winnipeg, Canada; Inflammatory Bowel Disease Clinical & Research Centre, University of Manitoba, Winnipeg, Canada; Children's Hospital Research Institute of Manitoba, Winnipeg, Canada.
| |
Collapse
|
2
|
Huang Y, Wang N, Ji X, Luo S, Gong L, Zhao C, Zheng G, Liu R, Zhang T. Apigenin ameliorates inflamed ulcerative colitis by regulating mast cell degranulation via the PAMP-MRGPRX2 feedback loop. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 140:156564. [PMID: 40054174 DOI: 10.1016/j.phymed.2025.156564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 02/10/2025] [Accepted: 02/23/2025] [Indexed: 03/25/2025]
Abstract
PURPOSE The aim of this study was to investigate the therapeutic effect of API on UC via the regulation of PAMP-MRGPRX2-mediated mast cells (MCs) degranulation. BACKGROUND The pro-inflammatory positive feedback loop mediated by Mas-related G-protein-coupled receptor X2 (MRGPRX2) and its endogenous ligand, PAMP-12, is associated with ulcerative colitis (UC) progression. However, the therapeutic strategies that target MRGPRX2 in the treatment of UC are less reported. Apigenin (API), a natural flavonoid, can relieve inflammation. METHOD A dextran sodium sulfate (DSS)-induced mouse UC model was used to elucidate the therapeutic effects of API. Animal behavior assessment, serological assays, and histological analysis were performed in wild-type (WT) and MC MrgprB2-conditional knockout (CKO) mouse model. mRNA sequencing analysis, PCR, ELISA, and western blotting were performed in vitro and in vivo to elucidate the mechanism underlying the effect of API by a PAMP-12 triggered MC degranulation model. RESULTS MC degranulation via MrgprB2 was critical for the persistence of inflammation in colitis. API attenuated colonic tissue damage, splenomegaly, and myeloperoxidase (MPO) activity in the colonic tissues. It also ameliorated colonic crypt structure damage and inflammatory cell infiltration. Moreover, API suppressed MCs degranulation, and the level of carboxypeptidases A3 (CPA3), in DSS-induced colitis, thereby blocking the pro-inflammatory positive feedback loop induced by PAMP-MrgprB2. Lastly, API effectively inhibited PAMP-12-triggered mast cell degranulation by regulating Akt1/XBP-1S/CHOP/TXNIP and NF-κB/IL-1β signaling pathways. CONCLUSION API alleviates inflammatory symptoms in UC by suppressing PAMP-MRGPRX2/B2 mediated MC sustained degranulation feedback loop.
Collapse
MESH Headings
- Animals
- Mast Cells/drug effects
- Colitis, Ulcerative/drug therapy
- Colitis, Ulcerative/chemically induced
- Colitis, Ulcerative/metabolism
- Receptors, G-Protein-Coupled/metabolism
- Receptors, G-Protein-Coupled/genetics
- Apigenin/pharmacology
- Cell Degranulation/drug effects
- Mice
- Mice, Knockout
- Mice, Inbred C57BL
- Disease Models, Animal
- Male
- Dextran Sulfate
- Receptors, Neuropeptide/metabolism
- Feedback, Physiological/drug effects
- Nerve Tissue Proteins/metabolism
- Signal Transduction/drug effects
Collapse
Affiliation(s)
- Yihan Huang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Na Wang
- Department of Otolaryngology, Affiliated Hospital of North China University of Science and Technology, Tangshan 063000, China
| | - Xiaolan Ji
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Shiqiong Luo
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Ling Gong
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Chenrui Zhao
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Guodong Zheng
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Rui Liu
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China.
| | - Tao Zhang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China.
| |
Collapse
|
3
|
Huyghe P, Ceulemans M, Keita ÅV, Söderholm J, Depoortere I, Tack J, Wauters L, Vanuytsel T. The Duodenal Microenvironment in Functional Dyspepsia. J Neurogastroenterol Motil 2025; 31:186-198. [PMID: 40205896 PMCID: PMC11986653 DOI: 10.5056/jnm24176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 02/20/2025] [Accepted: 02/22/2025] [Indexed: 04/11/2025] Open
Abstract
Functional dyspepsia (FD) is a chronic gastrointestinal disorder without a readily identifiable organic cause, resulting in bothersome upper abdominal symptoms. It is a highly prevalent disorder of which the pathophysiology remains mostly elusive, despite intensive research efforts. However, recent studies have found alterations in the microenvironment of the duodenum in patients with FD. In this review we summarize the duodenal microenvironment in homeostatic conditions and the alterations found in patients with FD, highlighting the similarities and discrepancies between different studies. The most consistent findings, being an impaired duodenal barrier and duodenal immune activation, are reviewed. We discuss the potential triggers for these observed alterations, including psychological comorbidities, luminal alterations and food related triggers. In summary, this review presents the evidence of molecular and cellular changes in patients with FD, with an impaired duodenal barrier and activated mucosal eosinophils and mast cells, challenging the notion that FD is purely functional, and offering different targets for potential future treatments.
Collapse
Affiliation(s)
- Pauline Huyghe
- Translational Research Centre for Gastrointestinal Disorders (TARGID), Department of Chronic Diseases and Metabolism (CHROMETA), Katholieke Universiteit Leuven, Leuven, Belgium
| | - Matthias Ceulemans
- Translational Research Centre for Gastrointestinal Disorders (TARGID), Department of Chronic Diseases and Metabolism (CHROMETA), Katholieke Universiteit Leuven, Leuven, Belgium
| | - Åsa V Keita
- Department of Biomedical and Clinical Sciences and Department of Surgery, Linköping University, Linköping, Sweden
| | - Johan Söderholm
- Department of Biomedical and Clinical Sciences and Department of Surgery, Linköping University, Linköping, Sweden
| | - Inge Depoortere
- Translational Research Centre for Gastrointestinal Disorders (TARGID), Department of Chronic Diseases and Metabolism (CHROMETA), Katholieke Universiteit Leuven, Leuven, Belgium
| | - Jan Tack
- Translational Research Centre for Gastrointestinal Disorders (TARGID), Department of Chronic Diseases and Metabolism (CHROMETA), Katholieke Universiteit Leuven, Leuven, Belgium
- Department of Gastroenterology and Hepatology, University Hospitals Leuven, Leuven, Belgium
| | - Lucas Wauters
- Translational Research Centre for Gastrointestinal Disorders (TARGID), Department of Chronic Diseases and Metabolism (CHROMETA), Katholieke Universiteit Leuven, Leuven, Belgium
- Department of Gastroenterology and Hepatology, University Hospitals Leuven, Leuven, Belgium
| | - Tim Vanuytsel
- Translational Research Centre for Gastrointestinal Disorders (TARGID), Department of Chronic Diseases and Metabolism (CHROMETA), Katholieke Universiteit Leuven, Leuven, Belgium
- Department of Gastroenterology and Hepatology, University Hospitals Leuven, Leuven, Belgium
| |
Collapse
|
4
|
Wei ZX, Jiang SH, Qi XY, Cheng YM, Liu Q, Hou XY, He J. scRNA-seq of the intestine reveals the key role of mast cells in early gut dysfunction associated with acute pancreatitis. World J Gastroenterol 2025; 31:103094. [PMID: 40182603 PMCID: PMC11962851 DOI: 10.3748/wjg.v31.i12.103094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Revised: 01/09/2025] [Accepted: 02/21/2025] [Indexed: 03/26/2025] Open
Abstract
BACKGROUND Intestinal barrier dysfunction is a prevalent and varied manifestation of acute pancreatitis (AP). Molecular mechanisms underlying the early intestinal barrier in AP remain poorly understood. AIM To explore the biological processes and mechanisms of intestinal injury associated with AP, and to find potential targets for early prevention or treatment of intestinal barrier injury. METHODS This study utilized single-cell RNA sequencing of the small intestine, alongside in vitro and in vivo experiments, to examine intestinal barrier function homeostasis during the early stages of AP and explore involved biological processes and potential mechanisms. RESULTS Seventeen major cell types and 33232 cells were identified across all samples, including normal, AP1 (4x caerulein injections, animals sacrificed 2 h after the last injection), and AP2 (8x caerulein injections, animals sacrificed 4 h after the last injection). An average of 980 genes per cell was found in the normal intestine, compared to 927 in the AP1 intestine and 1382 in the AP2 intestine. B cells, dendritic cells, mast cells (MCs), and monocytes in AP1 and AP2 showed reduced numbers compared to the normal intestine. Enterocytes, brush cells, enteroendocrine cells, and goblet cells maintained numbers similar to the normal intestine, while cytotoxic T cells and natural killer (NK) cells increased. Enterocytes in early AP exhibited elevated programmed cell death and intestinal barrier dysfunction but retained absorption capabilities. Cytotoxic T cells and NK cells showed enhanced pathogen-fighting abilities. Activated MCs, secreted chemokine (C-C motif) ligand 5 (CCL5), promoted neutrophil and macrophage infiltration and contributed to barrier dysfunction. CONCLUSION These findings enrich our understanding of biological processes and mechanisms in AP-associated intestinal injury, suggesting that CCL5 from MCs is a potential target for addressing dysfunction.
Collapse
Affiliation(s)
- Zu-Xing Wei
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan Province, China
| | - Shi-He Jiang
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan Province, China
| | - Xiao-Yan Qi
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan Province, China
| | - Yi-Miao Cheng
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan Province, China
| | - Qiong Liu
- Department of Stomatology, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan Province, China
| | - Xu-Yang Hou
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan Province, China
| | - Jun He
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan Province, China
| |
Collapse
|
5
|
Aljameeli AM, Alsuwayt B, Bharati D, Gohri V, Mohite P, Singh S, Chidrawar V. Chloride channels and mast cell function: pioneering new frontiers in IBD therapy. Mol Cell Biochem 2025:10.1007/s11010-025-05243-w. [PMID: 40038149 DOI: 10.1007/s11010-025-05243-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 02/22/2025] [Indexed: 03/06/2025]
Abstract
Emerging evidence indicates that chloride channels (ClCs) significantly affect the pathogenesis of inflammatory bowel disease (IBD) through their regulatory roles in mast cell function and epithelial integrity. IBD, encompassing conditions such as Crohn's disease and ulcerative colitis, involves chronic inflammation of the gastrointestinal tract, where channels influence immune responses, fluid balance, and cellular signalling pathways essential for maintaining mucosal homeostasis. This review examines the specific roles of ClC in mast cells, focussing on the regulation of mast cell activation, degranulation, cytokine release, and immune cell recruitment in inflamed tissues. Key channels, including Cystic Fibrosis Transmembrane Conductance Regulator (CFTR) and ClC-2, are discussed in detail because of their involvement in maintaining intestinal epithelial barrier function, a critical factor disrupted in IBD. For example, CFTR facilitates chloride ion transport across epithelial cells, which is essential for mucosal hydration and maintenance of the intestinal barrier. Reduced CFTR function can compromise this barrier, permitting microbial antigens to penetrate the underlying tissues and triggering excessive immune responses. ClC-2, another chloride channel expressed in mast cells and epithelial cells, supports tight junction integrity, contributes to barrier function, and reduces intestinal permeability. Dysregulation of these channels is linked to altered mast cell activity and excessive release of pro-inflammatory mediators, exacerbating IBD symptoms, such as diarrhoea, abdominal pain, and tissue damage. Here, we review recent pharmacological strategies targeting ClC, including CFTR potentiators and ClC-2 activators, which show the potential to mitigate inflammatory responses. Additionally, experimental approaches for selective modulation of chloride channels in mast cells have been explored. Although targeting ClC offers promising therapeutic avenues, challenges remain in achieving specificity and minimizing side effects. This review highlights the therapeutic potential of Cl channel modulation in mast cells as a novel approach for IBD treatment, aiming to reduce inflammation and restore intestinal homeostasis in affected patients.
Collapse
Affiliation(s)
- Ahmed M Aljameeli
- Department of Pharmacy Practice, College of Pharmacy, University of Hafr Al-Batin, Hafr Albatin, Saudi Arabia
| | - Bader Alsuwayt
- Department of Pharmacy Practice, College of Pharmacy, University of Hafr Al-Batin, Hafr Albatin, Saudi Arabia
| | - Deepak Bharati
- AETs St. John Institute of Pharmacy and Research, Palghar, Maharashtra, 401 404, India
| | - Vaishnavi Gohri
- AETs St. John Institute of Pharmacy and Research, Palghar, Maharashtra, 401 404, India
| | - Popat Mohite
- AETs St. John Institute of Pharmacy and Research, Palghar, Maharashtra, 401 404, India.
| | - Sudarshan Singh
- Office of Research Administration, Chiang Mai University, Chiang Mai, 50200, Thailand
- Faculty of Pharmacy, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Vijay Chidrawar
- School of Pharmacy and Technology Management, SVKM's Narsee Monjee Institute of Management Studies (NMIMS), Deemed-to-University, Green Industrial Park, TSIIC, Polepally, Jadcherla, Hyderabad, Telangana, 509301, India.
| |
Collapse
|
6
|
Meerschaert KA, Chiu IM. The gut-brain axis and pain signalling mechanisms in the gastrointestinal tract. Nat Rev Gastroenterol Hepatol 2025; 22:206-221. [PMID: 39578592 DOI: 10.1038/s41575-024-01017-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/25/2024] [Indexed: 11/24/2024]
Abstract
Visceral pain is a major clinical problem and one of the most common reasons patients with gastrointestinal disorders seek medical help. Peripheral sensory neurons that innervate the gut can detect noxious stimuli and send signals to the central nervous system that are perceived as pain. There is a bidirectional communication network between the gastrointestinal tract and the nervous system that mediates pain through the gut-brain axis. Sensory neurons detect mechanical and chemical stimuli within the intestinal tissues, and receive signals from immune cells, epithelial cells and the gut microbiota, which results in peripheral sensitization and visceral pain. This Review focuses on molecular communication between these non-neuronal cell types and neurons in visceral pain. These bidirectional interactions can be dysregulated during gastrointestinal diseases to exacerbate visceral pain. We outline the anatomical pathways involved in pain processing in the gut and how cell-cell communication is integrated into this gut-brain axis. Understanding how bidirectional communication between the gut and nervous system is altered during disease could provide new therapeutic targets for treating visceral pain.
Collapse
Affiliation(s)
| | - Isaac M Chiu
- Department of Immunology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
7
|
Sandberg-Janzon A, Karling P. Prescription of commonly used drugs in patients with functional bowel disorders. A cross-sectional comparison with the general population. Scand J Gastroenterol 2025; 60:253-261. [PMID: 39862135 DOI: 10.1080/00365521.2025.2458070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 01/13/2025] [Accepted: 01/19/2025] [Indexed: 01/27/2025]
Abstract
OBJECTIVES Comorbidity with other conditions is common in functional bowel disorders. We aimed to investigate the prescription patterns of commonly used drugs in patients with irritable bowel syndrome (IBS) and functional unspecific bowel disorder, compared to the general population. MATERIAL AND METHODS Prescriptions of commonly used drugs in 2022 were compared between patients and the general population from the same age group and region in Sweden. RESULTS Of 526 patients, 317 were followed up in 2022 (219 women and 98 men) and were compared to 51,001 women and 55,571 men in the general population. The median follow-up time from the first visit to 2022 was 8 years (25th-75th percentile 6-11 years). Female patients were significantly more likely than controls to be prescribed PPIs, antibiotics, NSAIDs, paracetamol, opioids, muscle relaxants, antimigraine drugs, antidepressants and asthma medications. Male patients were significantly more likely than controls to be prescribed PPIs, opioids, antidepressants, and asthma medications. In the year prior diagnosis and through 2022, female patients showed a significant decline in the use of PPIs (38% vs.10%; p < 0.001), antibiotics (27.5% vs. 20.1%; p = 0.0426), NSAIDs (23.3% vs.14.6%; p = 0.012), opioids (20.6% vs. 7.5%; p < 0.001), and a significantly increase in the use of asthma medications (15.5% vs. 24.2%; p = 0.0088). Male patients showed a significant decline in the use of PPIs and NSAIDs. CONCLUSION Patients with functional bowel disorders are more likely to be prescribed medications for conditions other than IBS. Over time, there was a decline in the prescriptions of most drugs, except for antidepressants and asthma medications.
Collapse
Affiliation(s)
| | - Pontus Karling
- Department of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
| |
Collapse
|
8
|
Grover M, Vanuytsel T, Chang L. Intestinal Permeability in Disorders of Gut-Brain Interaction: From Bench to Bedside. Gastroenterology 2025; 168:480-495. [PMID: 39236897 DOI: 10.1053/j.gastro.2024.08.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 06/27/2024] [Accepted: 08/20/2024] [Indexed: 09/07/2024]
Abstract
Intestinal barrier function lies at a critical interface of a range of peripheral and central processes that influence disorders of gut-brain interactions (DGBI). Although rigorously tested, the role of barrier dysfunction in driving clinical phenotype of DGBI remains to be fully elucidated. In vitro, in vivo, and ex vivo strategies can test various aspects of the broader permeability and barrier mechanisms in the gut. Luminal mediators of host, bacterial, and dietary origin can influence the barrier function and a disrupted barrier can also influence the luminal milieu. Critical to our understanding is how barrier dysfunction is influenced by stress and other comorbidities that associate with DGBI and the crosstalk between barrier and neural, hormonal, and immune responses. Additionally, the microbiome's significant role in the communication between the brain and gut has led to the integrative model of a microbiome gut-brain axis with reciprocal interactions between brain networks and networks composed of multiple cells in the gut, including immune cells, enterochromaffin cells, gut microbiota and the derived luminal mediators. This review highlights the techniques for assessment of barrier function, appraises evidence for barrier dysfunction in DGBI including mechanistic studies in humans, as well as provides an overview of therapeutic strategies that can be used to directly or indirectly restore barrier function in DGBI patients.
Collapse
Affiliation(s)
- Madhusudan Grover
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Tim Vanuytsel
- Translational Research Center for Gastrointestinal Disorders (TARGID), Department of Chronic Diseases and Metabolism (ChroMeta), KULeuven, Leuven, Belgium
| | - Lin Chang
- Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, University of California, Los Angeles, California.
| |
Collapse
|
9
|
Villablanca EJ. Organismal mucosal immunology: A perspective through the eyes of game theory. Mucosal Immunol 2025; 18:16-25. [PMID: 39672543 DOI: 10.1016/j.mucimm.2024.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 12/03/2024] [Accepted: 12/06/2024] [Indexed: 12/15/2024]
Abstract
In complex organisms, functional units must interact cohesively to maintain homeostasis, especially within mucosal barriers that house diverse, specialized cell exposed to constant environmental challenges. Understanding how homeostasis at mucosal barriers is maintained and how its disruption can lead to autoimmune diseases or cancer, requires a holistic view. Although omics approaches and systems immunology have become powerful tools, they are not without limitations; interpretations may reflect researchers' assumptions, even if other explanations exist. In this perspective, I propose that applying game theory concepts to mucosal immunology could help interpret complex data, offering fresh perspectives and supporting the exploration of alternative scenarios. By framing the mucosal immune system as a network of strategic interactions with multiple possible outcomes, game theory, which analyzes strategic interactions and decision-making processes, could illuminate novel cell types and functions, cell interactions, and responses to pathogens and commensals, leading to a more comprehensive understanding of immune homeostasis and diseases. In addition, game theory might encourage researchers to consider a broader range of possibilities, reduce the risk of myopic thinking, and ultimately enable a more refined and comprehensive understanding of the complexity of the immune system at mucosal barriers. This perspective aims to introduce game theory as a complementary framework for mucosal immunologists, encouraging them to incorporate these concepts into data interpretation and system modeling.
Collapse
Affiliation(s)
- Eduardo J Villablanca
- Division of Immunology and Respiratory Medicine, Department of Medicine Solna, Karolinska Institute and University Hospital, Stockholm, Sweden; Clinical Immunology and Transfusion Medicine, Karolinska University Hospital, Stockholm, Sweden; Center of Molecular Medicine, Stockholm, Sweden.
| |
Collapse
|
10
|
Pérez-Pons A, Henriques A, Contreras Sanfeliciano T, Jara-Acevedo M, Navarro-Navarro P, García-Montero AC, Álvarez-Twose I, Lecrevisse Q, Fluxa R, Sánchez-Muñoz L, Caldas C, Pozo J, González-López Ó, Pérez-Andrés M, Mayado A, Orfao A. Altered B-cell, plasma cell, and antibody immune profiles in blood of patients with systemic mastocytosis. J Allergy Clin Immunol 2025; 155:628-639. [PMID: 39423877 DOI: 10.1016/j.jaci.2024.10.005] [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: 04/30/2024] [Revised: 09/18/2024] [Accepted: 10/07/2024] [Indexed: 10/21/2024]
Abstract
BACKGROUND Systemic mastocytosis (SM) is a heterogeneous disease characterized by an expansion of KIT-mutated constitutively activated mast cells (MCs) that release MC mediators, which might act on the tumor microenvironment including other immune cells. OBJECTIVE To investigate the blood distribution of B-cell, plasma cell (PC), and antibody isotype compartments in patients with SM. METHODS We used spectral flow cytometry and the EuroFlow Immunomonitoring panel and Lymphocyte Screening Tube to quantify B cells, PCs, and their subsets in blood of 108 patients with SM (35 bone marrow mastocytosis [BMM] cases, 64 indolent SM [ISM] cases, 9 aggressive SM [ASM] cases) versus 117 age-matched healthy donors and paired bone marrow samples of 31 patients with SM versus 17 controls, respectively. In parallel, IgM, IgD, IgG, IgA, and IgE plasma levels were measured. RESULTS Compared with healthy donors, patients with SM showed increased immature B-cell production in bone marrow (P = .003) associated with greater release of pre-germinal center immature (P < .001) and naive CD5+ B lymphocytes (P < .001) to blood, but a pronounced decrease in PC counts of all different IgH isotypes and subclasses (P ≤ .001) together with overall increased IgM (P = .001) and IgD (P < .001) plasma levels. Different immune profiles were found per diagnostic subtype of disease with progressively greater counts in blood of immature B lymphocytes together with decreased IgMD+, IgG2+, IgA1+, and IgA2+ memory B cells (P ≤ .032) and elevated IgM (P = .017) plasma levels in cases of ASM, increased IgM (P = .001) and IgD (P = .001) plasma levels in ISM cases, and exacerbated IgE (P < .001) with decreased IgG (P = .008) plasma levels in BMM cases. CONCLUSIONS Our results reveal a significant dysregulation of the B-cell and PC compartments in blood of patients with SM, consistent with distinctly altered antibody isotype profiles in plasma of patients with BMM versus ISM versus ASM.
Collapse
Affiliation(s)
- Alba Pérez-Pons
- Cancer Research Center, Department of Medicine and Cytometry Service, University of Salamanca, Salamanca, Spain; Biomedical Research Institute of Salamanca, Salamanca, Spain; Biomedical Research Networking Center Consortium, Madrid, Spain; Spanish Network on Mastocytosis, Toledo and Salamanca, Spain
| | - Ana Henriques
- Spanish Network on Mastocytosis, Toledo and Salamanca, Spain; Instituto de Estudios de Mastocitosis de Castilla La Mancha, Virgen del Valle Hospital, Toledo and Madrid, Spain
| | | | - María Jara-Acevedo
- Cancer Research Center, Department of Medicine and Cytometry Service, University of Salamanca, Salamanca, Spain; Biomedical Research Institute of Salamanca, Salamanca, Spain; Biomedical Research Networking Center Consortium, Madrid, Spain; Spanish Network on Mastocytosis, Toledo and Salamanca, Spain; Sequencing Service, University of Salamanca, Salamanca, Spain
| | - Paula Navarro-Navarro
- Cancer Research Center, Department of Medicine and Cytometry Service, University of Salamanca, Salamanca, Spain; Biomedical Research Institute of Salamanca, Salamanca, Spain; Biomedical Research Networking Center Consortium, Madrid, Spain; Spanish Network on Mastocytosis, Toledo and Salamanca, Spain; Sequencing Service, University of Salamanca, Salamanca, Spain
| | - Andrés C García-Montero
- Cancer Research Center, Department of Medicine and Cytometry Service, University of Salamanca, Salamanca, Spain; Biomedical Research Institute of Salamanca, Salamanca, Spain; Biomedical Research Networking Center Consortium, Madrid, Spain; Spanish Network on Mastocytosis, Toledo and Salamanca, Spain
| | - Iván Álvarez-Twose
- Biomedical Research Networking Center Consortium, Madrid, Spain; Spanish Network on Mastocytosis, Toledo and Salamanca, Spain; Instituto de Estudios de Mastocitosis de Castilla La Mancha, Virgen del Valle Hospital, Toledo and Madrid, Spain
| | - Quentin Lecrevisse
- Cancer Research Center, Department of Medicine and Cytometry Service, University of Salamanca, Salamanca, Spain; Biomedical Research Institute of Salamanca, Salamanca, Spain; Biomedical Research Networking Center Consortium, Madrid, Spain
| | | | - Laura Sánchez-Muñoz
- Spanish Network on Mastocytosis, Toledo and Salamanca, Spain; Instituto de Estudios de Mastocitosis de Castilla La Mancha, Virgen del Valle Hospital, Toledo and Madrid, Spain
| | - Carolina Caldas
- Cancer Research Center, Department of Medicine and Cytometry Service, University of Salamanca, Salamanca, Spain; Biomedical Research Institute of Salamanca, Salamanca, Spain; Spanish Network on Mastocytosis, Toledo and Salamanca, Spain
| | - Julio Pozo
- Cancer Research Center, Department of Medicine and Cytometry Service, University of Salamanca, Salamanca, Spain; Biomedical Research Institute of Salamanca, Salamanca, Spain
| | - Óscar González-López
- Cancer Research Center, Department of Medicine and Cytometry Service, University of Salamanca, Salamanca, Spain; Biomedical Research Institute of Salamanca, Salamanca, Spain; Spanish Network on Mastocytosis, Toledo and Salamanca, Spain
| | - Martín Pérez-Andrés
- Cancer Research Center, Department of Medicine and Cytometry Service, University of Salamanca, Salamanca, Spain; Biomedical Research Institute of Salamanca, Salamanca, Spain; Biomedical Research Networking Center Consortium, Madrid, Spain
| | - Andrea Mayado
- Cancer Research Center, Department of Medicine and Cytometry Service, University of Salamanca, Salamanca, Spain; Biomedical Research Institute of Salamanca, Salamanca, Spain; Biomedical Research Networking Center Consortium, Madrid, Spain; Spanish Network on Mastocytosis, Toledo and Salamanca, Spain
| | - Alberto Orfao
- Cancer Research Center, Department of Medicine and Cytometry Service, University of Salamanca, Salamanca, Spain; Biomedical Research Institute of Salamanca, Salamanca, Spain; Biomedical Research Networking Center Consortium, Madrid, Spain; Spanish Network on Mastocytosis, Toledo and Salamanca, Spain.
| |
Collapse
|
11
|
Zhou M, Lv J, Chen X, Shi Y, Chao G, Zhang S. From gut to liver: Exploring the crosstalk between gut-liver axis and oxidative stress in metabolic dysfunction-associated steatotic liver disease. Ann Hepatol 2025; 30:101777. [PMID: 39832564 DOI: 10.1016/j.aohep.2025.101777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 01/05/2025] [Accepted: 01/10/2025] [Indexed: 01/22/2025]
Abstract
Non-alcoholic fatty liver disease (NAFLD), now recognized as metabolic dysfunction-associated steatotic liver disease (MASLD), represents a significant and escalating global health challenge. Its prevalence is intricately linked to obesity, insulin resistance, and other components of the metabolic syndrome. As our comprehension of MASLD deepens, it has become evident that this condition extends beyond the liver, embodying a complex, multi-systemic disease with hepatic manifestations that mirror the broader metabolic landscape. This comprehensive review delves into the critical interplay between the gut-liver axis and oxidative stress, elucidating their pivotal roles in the etiology and progression of MASLD. Our analysis reveals several key findings: (1) Bile acid dysregulation can trigger oxidative stress through enhanced ROS production in hepatocytes and Kupffer cells, leading to mitochondrial dysfunction and lipid peroxidation; (2) Gut microbiota dysbiosis disrupts intestinal barrier function, allowing increased translocation of endotoxins like LPS, which activate inflammatory pathways through TLR4 signaling and promote oxidative stress via NADPH oxidase activation; (3) The redox-sensitive transcription factors NF-κB and Nrf2 serve as crucial mediators in the gut-liver axis, with NF-κB regulating inflammatory responses and Nrf2 orchestrating antioxidant defenses; (4) Oxidative stress-induced damage to intestinal barrier function creates a destructive feedback loop, further exacerbating liver inflammation and disease progression. These findings highlight the complex interrelationship between gut-liver axis dysfunction and oxidative stress in MASLD pathogenesis, suggesting potential therapeutic targets for disease management.
Collapse
Affiliation(s)
- Mi Zhou
- Department of Gastroenterology, Xinhua Hospital of zhejiang Province: The Second Affiliated Hospital of Zhejiang Chinese Medical University, Zhejiang, China
| | - Jianyu Lv
- Department of Gastroenterology, Xinhua Hospital of zhejiang Province: The Second Affiliated Hospital of Zhejiang Chinese Medical University, Zhejiang, China
| | - Xinli Chen
- Department of Gastroenterology, Xinhua Hospital of zhejiang Province: The Second Affiliated Hospital of Zhejiang Chinese Medical University, Zhejiang, China
| | - Yujie Shi
- Department of Gastroenterology, Xinhua Hospital of zhejiang Province: The Second Affiliated Hospital of Zhejiang Chinese Medical University, Zhejiang, China
| | - Guanqun Chao
- Department of General Practice, Zhejiang University School of Medicine Sir Run Shaw Hospital, China
| | - Shuo Zhang
- Department of Gastroenterology, Xinhua Hospital of zhejiang Province: The Second Affiliated Hospital of Zhejiang Chinese Medical University, Zhejiang, China.
| |
Collapse
|
12
|
Li J, Xiao C, Li C, He J. Tissue-resident immune cells: from defining characteristics to roles in diseases. Signal Transduct Target Ther 2025; 10:12. [PMID: 39820040 PMCID: PMC11755756 DOI: 10.1038/s41392-024-02050-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 09/28/2024] [Accepted: 11/04/2024] [Indexed: 01/19/2025] Open
Abstract
Tissue-resident immune cells (TRICs) are a highly heterogeneous and plastic subpopulation of immune cells that reside in lymphoid or peripheral tissues without recirculation. These cells are endowed with notably distinct capabilities, setting them apart from their circulating leukocyte counterparts. Many studies demonstrate their complex roles in both health and disease, involving the regulation of homeostasis, protection, and destruction. The advancement of tissue-resolution technologies, such as single-cell sequencing and spatiotemporal omics, provides deeper insights into the cell morphology, characteristic markers, and dynamic transcriptional profiles of TRICs. Currently, the reported TRIC population includes tissue-resident T cells, tissue-resident memory B (BRM) cells, tissue-resident innate lymphocytes, tissue-resident macrophages, tissue-resident neutrophils (TRNs), and tissue-resident mast cells, but unignorably the existence of TRNs is controversial. Previous studies focus on one of them in specific tissues or diseases, however, the origins, developmental trajectories, and intercellular cross-talks of every TRIC type are not fully summarized. In addition, a systemic overview of TRICs in disease progression and the development of parallel therapeutic strategies is lacking. Here, we describe the development and function characteristics of all TRIC types and their major roles in health and diseases. We shed light on how to harness TRICs to offer new therapeutic targets and present burning questions in this field.
Collapse
Affiliation(s)
- Jia Li
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chu Xiao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chunxiang Li
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Jie He
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
13
|
Céspedes N, Tsolis RM, Piliponsky AM, Luckhart S. The type 2 immune response in gut homeostasis and parasite transmission in malaria. Trends Parasitol 2025; 41:38-51. [PMID: 39658487 DOI: 10.1016/j.pt.2024.11.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 11/18/2024] [Accepted: 11/18/2024] [Indexed: 12/12/2024]
Abstract
Malaria predisposes to concomitant bacteremia, resulting in increased mortality risk. Previous studies indicated that malaria causes structural changes in the intestine, induces tolerogenic immune responses, inhibits neutrophil recruitment, suppresses innate synthesis of IFN-γ, dysregulates mast cells (MCs) and basophils, and induces Th2-type immune responses. These can reduce parasite control while increasing enteropathogenic dissemination. Moreover, there is growing evidence that Th2 immunity, while protecting the host from overwhelming inflammation, may also contribute to increased parasite transmission. This review explores the roles of the regulatory immune response in bacterial coinfections and parasite transmission in malaria.
Collapse
Affiliation(s)
- Nora Céspedes
- Department of Entomology, Plant Pathology and Nematology, University of Idaho, Moscow, ID, USA.
| | - Renée M Tsolis
- Department of Medical Microbiology and Immunology, University of California-Davis, Davis, CA, USA
| | - Adrian M Piliponsky
- Department of Pediatrics and Department of Pathology, Seattle Children's Research Institute, Seattle, WA, USA
| | - Shirley Luckhart
- Department of Entomology, Plant Pathology and Nematology, University of Idaho, Moscow, ID, USA; Department of Biological Sciences, University of Idaho, Moscow, ID, USA
| |
Collapse
|
14
|
Chen S, Jiang D, Zhuang Q, Hou X, Jia X, Chen J, Lin H, Zhang M, Tan N, Xiao Y. Esophageal microbial dysbiosis impairs mucosal barrier integrity via toll-like receptor 2 pathway in patients with gastroesophageal reflux symptoms. J Transl Med 2024; 22:1145. [PMID: 39719586 DOI: 10.1186/s12967-024-05878-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 11/11/2024] [Indexed: 12/26/2024] Open
Abstract
BACKGROUND Previous research on the lower gastrointestinal tract has proved that microbial dysbiosis can lead to intestinal barrier dysfunction and enhanced visceral sensitivity, thus triggering bowel symptoms. Whether esophageal microbial dysbiosis also contributes to the development of gastroesophageal reflux (GER) symptoms, which are known to be associated with impaired esophageal barrier integrity, remains to be explored. METHODS Patients with GER symptoms (gastroesophageal reflux disease [GERD] and functional esophageal disorders [FED]), duodenal ulcer patients and healthy controls were prospectively included for esophageal microbial analysis. The expression of toll-like receptors (TLRs) and tight junction proteins and intercellular spaces were assessed through transcriptome analysis and immunohistochemistry. The human esophageal epithelial cell (HEEC) line was used to explore how esophageal microbial dysbiosis induced GER symptoms. RESULTS Patients with GER symptoms, whether GERD or FED, had a very similar pattern of microbial composition, which showed a significantly increased proportion of Gram-negative bacteria than controls. Patients with GER symptoms (GERD and FED) also exhibited significantly higher TLR2 expression, reduced claudin-1 expression and dilated intercellular spaces (DIS). In vitro, exposure of HEECs to lipopolysaccharide resulted in marked up-regulation of TLR2 and interleukin (IL)-6, down-regulation of claudin-1 and DIS. These effects were mitigated by blocking TLR2 or IL-6. CONCLUSION This study demonstrated that regardless of objective evidence of reflux, patients with GER symptoms presented esophageal microbial dysbiosis characterized by an elevated proportion of Gram-negative bacteria. Enriched Gram-negative bacteria could induce esophageal barrier dysfunction via LPS-TLR2-IL-6-claudin-1-DIS pathway.
Collapse
Affiliation(s)
- Songfeng Chen
- Department of Gastroenterology, The First Affiliated Hospital of Sun Yat-sen University, No. 58 Zhongshan Road 2, Guangzhou, Guangdong, 510080, China
| | - Dianxuan Jiang
- Department of Gastroenterology, The First Affiliated Hospital of Sun Yat-sen University, No. 58 Zhongshan Road 2, Guangzhou, Guangdong, 510080, China
| | - Qianjun Zhuang
- Department of Gastroenterology, The First Affiliated Hospital of Sun Yat-sen University, No. 58 Zhongshan Road 2, Guangzhou, Guangdong, 510080, China
| | - Xun Hou
- Gastrointestinal Surgery Center, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xingyu Jia
- Department of Gastroenterology, The First Affiliated Hospital of Sun Yat-sen University, No. 58 Zhongshan Road 2, Guangzhou, Guangdong, 510080, China
| | - Jing Chen
- Department of Gastroenterology, The First Affiliated Hospital of Sun Yat-sen University, No. 58 Zhongshan Road 2, Guangzhou, Guangdong, 510080, China
| | - Huiting Lin
- Department of Gastroenterology, The First Affiliated Hospital of Sun Yat-sen University, No. 58 Zhongshan Road 2, Guangzhou, Guangdong, 510080, China
| | - Mengyu Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Sun Yat-sen University, No. 58 Zhongshan Road 2, Guangzhou, Guangdong, 510080, China
| | - Niandi Tan
- Department of Gastroenterology, The First Affiliated Hospital of Sun Yat-sen University, No. 58 Zhongshan Road 2, Guangzhou, Guangdong, 510080, China
| | - Yinglian Xiao
- Department of Gastroenterology, The First Affiliated Hospital of Sun Yat-sen University, No. 58 Zhongshan Road 2, Guangzhou, Guangdong, 510080, China.
| |
Collapse
|
15
|
Aguiar A, Menezes de Brito ASS, Santos AGAD, Watanabe PDS, Cuman RKN, Trevizan AR, de Lima LL, Bersani-Amado CA, Rinaldi JDC, Sant Ana DDMG, Nogueira-Melo GDA. Mastocytosis and intraepithelial lymphocytosis in the ileum and colon characterize chronic Toxoplasma gondii infection in mice. Tissue Cell 2024; 91:102533. [PMID: 39213782 DOI: 10.1016/j.tice.2024.102533] [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: 02/08/2024] [Revised: 08/13/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024]
Abstract
Toxoplasma gondii is the causative agent of toxoplasmosis, a common zoonotic disease affecting vertebrates with high global incidence. For the parasite to disseminate throughout the body, it crosses the intestinal barrier, triggering inflammatory reactions. This study aimed to assess the tissue response in the ileum and colon of mice following chronic infection with T. gondii. Fourteen mice were divided into two groups: the infected group received 1000 T. gondii oocysts via gavage, and after 60 days, the mice were euthanized. The ileum and colon were collected and processed for histological analysis, inflammatory marker measurement and myenteric neuron analysis. Chronic infection resulted in a significant increase in intraepithelial lymphocytes and mast cells, as well as morphometric changes such as increased total intestinal wall thickness of the ileum, crypt depth, collagen fiber area, and a decrease in myeloperoxidase activity, without altering nitric oxide levels. While the number of myenteric neurons remained unchanged, there was an increase in vasoactive intestinal peptide expression. These results suggest persistence intestinal inflammatory stimuli in chronic T. gondii infection.
Collapse
Affiliation(s)
- Aline Aguiar
- Graduate Program in Biosciences and Pathophysiology, State University of Maringá, Maringá, Paraná, Brazil
| | | | | | - Paulo da Silva Watanabe
- Graduate Program in Biosciences and Pathophysiology, State University of Maringá, Maringá, Paraná, Brazil
| | | | - Aline Rosa Trevizan
- Graduate Program in Biosciences and Pathophysiology, State University of Maringá, Maringá, Paraná, Brazil
| | - Lainy Leiny de Lima
- Graduate Program in Biosciences and Pathophysiology, State University of Maringá, Maringá, Paraná, Brazil
| | | | | | | | | |
Collapse
|
16
|
Liu Z, Wang H, Han H, Li N, Zheng Z, Liang S, Zhong R, Chen L, Yan J, Mu S. The protective effect of dulcitol on lipopolysaccharide-induced intestinal injury in piglets: mechanistic insights. J Nutr Biochem 2024; 133:109719. [PMID: 39103108 DOI: 10.1016/j.jnutbio.2024.109719] [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: 05/07/2024] [Revised: 07/29/2024] [Accepted: 07/31/2024] [Indexed: 08/07/2024]
Abstract
This study investigated the protective effect of dulcitol on LPS-induced intestinal injury in piglets and explored the underlying molecular mechanisms. A total of 108 piglets were divided into three groups: CON, LPS, and DUL. The CON and LPS groups were fed a basal diet, the DUL group was fed a diet supplementation with 500 mg/kg dulcitol. On day 29, 6 piglets in the LPS and DUL groups were injected with 100 μg/kg BW of LPS. At 4 h postchallenge, all pigs were slaughtered, and colonic samples were collected. Results showed that dulcitol supplementation boosted intestinal barrier function in LPS-challenged piglets by enhancing intestinal morphology and integrity, and increasing the gene expression of zonula occludens-1, claudin-1, and occludin in the colonic mucosa (P <0.05). Metabolomics showed DUL supplementation mainly increased (P <0.05) the metabolites related to steroid and vitamin metabolism (Cholesterol and Vitamin C). Proteomics showed that dulcitol supplementation altered the protein expression involved in maintaining barrier integrity (FN1, CADM1, and PARD3), inhibiting inflammatory response (SLP1, SFN, and IRF3), and apoptosis (including FAS, ING1, BTK, MTHFR, NOX, and P53BP2) in LPS-challenged piglets (P <0.05). Additionally, dulcitol addition also suppressed the TLR4/NF-κB signaling pathway and apoptosis in mRNA and protein levels. Dulcitol increased the abundance of short-chain fatty acid-producing bacteria (Lactobacillus, Blautia, and Faecalibacterium) at the genus level, but decreased the relative abundance of Proteobacteria at the phylum level and Pseudomonas and Delftia at the genus level in piglets (P<.05). In conclusion, these results suggested that the addition of dulcitol alleviated LPS-induced intestinal barrier injury in piglets, probably by maintaining its integrity, inhibiting the TLR4/NF-κB signaling pathways and apoptosis, and modulating the gut microbiota. Therefore, dulcitol can be considered a potential dietary additive for improving intestinal health in pig models.
Collapse
Affiliation(s)
- Zhengqun Liu
- Tianjin Key Laboratory of Animal Molecular Breeding and Biotechnology; Tianjin Engineering Research Center of Animal Healthy Farming; Institute of Animal Science and Veterinary, Tianjin Academy of Agricultural Sciences, Tianjin, China; State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China; Tianfu Institute of Research and Innovation, Southwest University of Science and Technology, Chengdu, Sichuan, China
| | - Han Wang
- Tianjin Key Laboratory of Animal Molecular Breeding and Biotechnology; Tianjin Engineering Research Center of Animal Healthy Farming; Institute of Animal Science and Veterinary, Tianjin Academy of Agricultural Sciences, Tianjin, China; College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Hui Han
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Ning Li
- Tianjin Key Laboratory of Animal Molecular Breeding and Biotechnology; Tianjin Engineering Research Center of Animal Healthy Farming; Institute of Animal Science and Veterinary, Tianjin Academy of Agricultural Sciences, Tianjin, China
| | - Zi Zheng
- Tianjin Key Laboratory of Animal Molecular Breeding and Biotechnology; Tianjin Engineering Research Center of Animal Healthy Farming; Institute of Animal Science and Veterinary, Tianjin Academy of Agricultural Sciences, Tianjin, China
| | - Shiyue Liang
- Tianjin Key Laboratory of Animal Molecular Breeding and Biotechnology; Tianjin Engineering Research Center of Animal Healthy Farming; Institute of Animal Science and Veterinary, Tianjin Academy of Agricultural Sciences, Tianjin, China
| | - Ruqing Zhong
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Liang Chen
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Jun Yan
- Tianjin Key Laboratory of Animal Molecular Breeding and Biotechnology; Tianjin Engineering Research Center of Animal Healthy Farming; Institute of Animal Science and Veterinary, Tianjin Academy of Agricultural Sciences, Tianjin, China.
| | - Shuqin Mu
- Tianjin Key Laboratory of Animal Molecular Breeding and Biotechnology; Tianjin Engineering Research Center of Animal Healthy Farming; Institute of Animal Science and Veterinary, Tianjin Academy of Agricultural Sciences, Tianjin, China.
| |
Collapse
|
17
|
Paulraj RS, Afroz S, Palaniappan B, Murughiyan U, Singh S, Arthur S, Sundaram U. Intestinal Epithelial Cell Brush Border Membrane Cl:HCO 3 Exchanger Regulation by Mast Cells in Chronic Ileitis. Int J Mol Sci 2024; 25:11208. [PMID: 39456989 PMCID: PMC11508389 DOI: 10.3390/ijms252011208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 09/30/2024] [Accepted: 10/06/2024] [Indexed: 10/28/2024] Open
Abstract
Malabsorption of NaCl is the primary cause of diarrhea in inflammatory bowel disease (IBD). Coupled NaCl absorption occurs via the dual operation of Na:H and Cl:HCO3 exchange in the brush border membrane (BBM) of villus cells. Cl:HCO3 exchange is mediated by BBM transporters DRA (downregulated in adenoma) and PAT1 (putative anion transporter 1) in the mammalian small intestine. DRA/PAT1-mediated Cl:HCO3 exchange was significantly downregulated in the BBM of villus cells in a rabbit model of chronic ileitis, while Na:H exchange was unaffected. The inhibition of Cl:HCO3 exchange was restored in the rabbits when treated with a broad-spectrum immunomodulator, i.e. a glucocorticoid, indicating that the downregulation of DRA/PAT1 is likely to be immune-mediated during chronic enteritis. Mucosal mast cells are one type of key immune cells that are known to proliferate and release immune inflammatory mediators, thus playing a significant role in the pathogenesis of IBD. However, how mast cells may regulate DRA- and PAT1-mediated Cl:HCO3 exchange in a rabbit model of chronic ileitis is unknown. In this study, treatment of rabbits with chronic intestinal inflammation with the mast cell stabilizer ketotifen did not affect the mucosal architecture of the inflamed intestine. However, ketotifen treatment reversed the inhibition of Cl:HCO3 activity in the BBM of villus cells. This restoration of Cl:HCO3 activity to normal levels by ketotifen was found to be secondary to restoring the affinity of the exchangers for its substrate chloride. This observation was consistent with molecular studies, where the mRNA and BBM protein expressions of DRA and PAT1 remained unaffected in the villus cells under all experimental conditions. Thus, this study indicates that mast cells mediated the inhibition of coupled NaCl absorption by inhibiting Cl:HCO3 exchange in a rabbit model of chronic enteritis.
Collapse
Affiliation(s)
- Raja Singh Paulraj
- Department of Clinical and Translational Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25701, USA (B.P.); (U.M.)
| | - Sheuli Afroz
- Department of Clinical and Translational Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25701, USA (B.P.); (U.M.)
| | - Balasubramanian Palaniappan
- Department of Clinical and Translational Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25701, USA (B.P.); (U.M.)
| | - Usha Murughiyan
- Department of Clinical and Translational Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25701, USA (B.P.); (U.M.)
- Department of Internal Medicine, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25701, USA
| | - Soudamani Singh
- Department of Clinical and Translational Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25701, USA (B.P.); (U.M.)
| | - Subha Arthur
- Department of Clinical and Translational Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25701, USA (B.P.); (U.M.)
| | - Uma Sundaram
- Department of Clinical and Translational Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25701, USA (B.P.); (U.M.)
| |
Collapse
|
18
|
Van Remoortel S, Hussein H, Boeckxstaens G. Mast cell modulation: A novel therapeutic strategy for abdominal pain in irritable bowel syndrome. Cell Rep Med 2024; 5:101780. [PMID: 39378882 PMCID: PMC11513802 DOI: 10.1016/j.xcrm.2024.101780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 07/10/2024] [Accepted: 09/17/2024] [Indexed: 10/10/2024]
Abstract
Irritable bowel syndrome (IBS) is one of the most prevalent gastrointestinal disorders characterized by recurrent abdominal pain and an altered defecation pattern. Chronic abdominal pain represents the hallmark IBS symptom and is reported to have the most bothersome impact on the patient's quality of life. Unfortunately, effective therapeutic strategies reducing abdominal pain are lacking, mainly attributed to a limited understanding of the contributing mechanisms. In the past few years, exciting new insights have pointed out that altered communication between gut immune cells and pain-sensing nerves acts as a hallmark driver of IBS-related abdominal pain. In this review, we aim to summarize our current knowledge on altered neuro-immune crosstalk as the main driver of altered pain signaling, with a specific focus on altered mast cell functioning herein, and highlight the relevance of targeting mast cell-mediated mechanisms as a novel therapeutic strategy for chronic abdominal pain in IBS patients.
Collapse
Affiliation(s)
- Samuel Van Remoortel
- Translational Research Centre for Gastrointestinal Disorders, KU Leuven, Leuven, Belgium
| | - Hind Hussein
- Translational Research Centre for Gastrointestinal Disorders, KU Leuven, Leuven, Belgium
| | - Guy Boeckxstaens
- Translational Research Centre for Gastrointestinal Disorders, KU Leuven, Leuven, Belgium.
| |
Collapse
|
19
|
Li FY, Wu X, Yao MF, Zhang J, Mo YJ. Identification and validation of coagulation and fibrinolytic-related diagnostic biomarkers for ulcerative colitis by bioinformatics analysis. Medicine (Baltimore) 2024; 103:e39552. [PMID: 39252244 PMCID: PMC11383730 DOI: 10.1097/md.0000000000039552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 08/13/2024] [Indexed: 09/11/2024] Open
Abstract
Abnormalities in coagulation and fibrinolytic status have been demonstrated to be relevant to inflammatory bowel disease. Nevertheless, there is no study to methodically examine the role of the coagulation and fibrinolysis-related genes in the diagnosis of ulcerative colitis (UC). UC-related datasets (GSE169568 and GSE94648) were originated from the Gene Expression Omnibus database. The biomarkers related to coagulation and fibrinolysis were identified through combining differentially expressed analysis and machine learning algorithms. Moreover, Gene Set Enrichment Analysis and immune analysis were carried out. A total of 4 biomarkers (MAP2K1, CREBBP, TAF1, and HP) were identified, and biomarkers were markedly enriched in pathways related to immunity, such as T-cell receptor signaling pathway, primary immunodeficiency, chemokine signaling pathway, etc. In total, the infiltrating abundance of 4 immune cells between UC and control was markedly different, namely eosinophils, macrophage M0, resting mast cells, and regulatory T cells. And all biomarkers were significantly relevant to eosinophils. Our findings detected 4 coagulation and fibrinolysis-related biomarkers (MAP2K1, CREBBP, TAF1, and HP) for UC, which contributed to the advancement of UC for further clinical investigation.
Collapse
Affiliation(s)
- Feng-Yun Li
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou Province, China
| | - Xue Wu
- Department of Proctology, The First People’s Hospital of Bijie, Bijie City, Guizhou Province, China
| | - Mei-Fang Yao
- The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou Province, China
| | - Juan Zhang
- Renhuai Hospital of Traditional Chinese Medicine, Renhuai City, Guizhou Province, China
| | - Yuan-Jun Mo
- Renhuai Hospital of Traditional Chinese Medicine, Renhuai City, Guizhou Province, China
| |
Collapse
|
20
|
Hussein H, Van Remoortel S, Boeckxstaens GE. Irritable bowel syndrome: When food is a pain in the gut. Immunol Rev 2024; 326:102-116. [PMID: 39037230 DOI: 10.1111/imr.13374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/23/2024]
Abstract
Irritable bowel syndrome (IBS) is a chronic gastrointestinal condition associated with altered bowel habits and recurrent abdominal pain, often triggered by food intake. Current treatments focus on improving stool pattern, but effective treatments for pain in IBS are still lacking due to our limited understanding of pathophysiological mechanisms. Visceral hypersensitivity (VHS), or abnormal visceral pain perception, underlies abdominal pain development in IBS, and mast cell activation has been shown to play an important role in the development of VHS. Our work recently revealed that abdominal pain in response to food intake is induced by the sensitization of colonic pain-sensing neurons by histamine produced by activated mast cells following a local IgE response to food. In this review, we summarize the current knowledge on abdominal pain and VHS pathophysiology in IBS, we outline the work leading to the discovery of the role of histamine in abdominal pain, and we introduce antihistamines as a novel treatment option to manage chronic abdominal pain in patients with IBS.
Collapse
Affiliation(s)
- Hind Hussein
- Center for Intestinal Neuro-Immune Interactions, Translational Research in Gastrointestinal Disorders (TARGID), Department of Chronic Diseases, Metabolism, and Ageing (CHROMETA), KU Leuven, Leuven, Belgium
| | - Samuel Van Remoortel
- Center for Intestinal Neuro-Immune Interactions, Translational Research in Gastrointestinal Disorders (TARGID), Department of Chronic Diseases, Metabolism, and Ageing (CHROMETA), KU Leuven, Leuven, Belgium
| | - Guy E Boeckxstaens
- Center for Intestinal Neuro-Immune Interactions, Translational Research in Gastrointestinal Disorders (TARGID), Department of Chronic Diseases, Metabolism, and Ageing (CHROMETA), KU Leuven, Leuven, Belgium
| |
Collapse
|
21
|
Nair B, Kamath AJ, Tergaonkar V, Sethi G, Nath LR. Mast cells and the gut-liver Axis: Implications for liver disease progression and therapy. Life Sci 2024; 351:122818. [PMID: 38866220 DOI: 10.1016/j.lfs.2024.122818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 05/24/2024] [Accepted: 06/07/2024] [Indexed: 06/14/2024]
Abstract
The role of mast cells, traditionally recognized for their involvement in immediate hypersensitivity reactions, has garnered significant attention in liver diseases. Studies have indicated a notable increase in mast cell counts following hepatic injury, underscoring their potential contribution to liver disorder pathogenesis. Predominantly situated in connective tissue that envelops the hepatic veins, bile ducts, and arteries, mast cells are central to both initiating and perpetuating liver disorders. Additionally, they are crucial for maintaining gastrointestinal barrier function. The gut-liver axis emphasizes the complex, two-way communication between the gut microbiome and the liver. Past research has implicated gut microbiota and their metabolites in the progression of hepatic disorders. This review sheds light on how mast cells are activated in various liver conditions such as alcoholic liver disease (ALD), non-alcoholic fatty liver disease (NAFLD), viral hepatitis, hepatic fibrogenesis, and hepatocellular carcinoma. It also briefly explores the connection between the gut microbiome and mast cell activation in these hepatic conditions.
Collapse
Affiliation(s)
- Bhagyalakshmi Nair
- Department of Pharmacognosy, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Ponekkara, P.O., Kochi, Kerala 682041, India; Department of Pharmacology, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Ponekkara, P.O., Kochi, Kerala 682041, India
| | - Adithya Jayaprakash Kamath
- Department of Pharmacognosy, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Ponekkara, P.O., Kochi, Kerala 682041, India; Department of Pharmaceutics, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Ponekkara, P.O., Kochi, Kerala 682041, India
| | - Vinay Tergaonkar
- Laboratory of NFκB Signalling, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, 138673, Singapore
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 117600, Singapore.
| | - Lekshmi R Nath
- Department of Pharmacognosy, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Ponekkara, P.O., Kochi, Kerala 682041, India.
| |
Collapse
|
22
|
Sait AM, Day PJR. Interconnections between the Gut Microbiome and Alzheimer's Disease: Mechanisms and Therapeutic Potential. Int J Mol Sci 2024; 25:8619. [PMID: 39201303 PMCID: PMC11354889 DOI: 10.3390/ijms25168619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 07/23/2024] [Accepted: 08/06/2024] [Indexed: 09/02/2024] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease that is known to accumulate amyloid-β (Aβ) and tau protein. Clinical studies have not identified pathogenesis mechanisms or produced an effective cure for AD. The Aβ monoclonal antibody lecanemab reduces Aβ plaque formation for the treatment of AD, but more studies are required to increase the effectiveness of drugs to reduce cognitive decline. The lack of AD therapy targets and evidence of an association with an acute neuroinflammatory response caused by several bacteria and viruses in some individuals has led to the establishment of the infection hypothesis during the last 10 years. How pathogens cross the blood-brain barrier is highly topical and is seen to be pivotal in proving the hypothesis. This review summarizes the possible role of the gut microbiome in the pathogenesis of AD and feasible therapeutic approaches and current research limitations.
Collapse
Affiliation(s)
- Ahmad M. Sait
- Medical Laboratory Science, Faculty of Applied Medical Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
- Regenerative Medicine Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Philip J. R. Day
- Division of Evolution and Genomic Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PL, UK
- Department of Medicine, University of Cape Town, Cape Town 7925, South Africa
| |
Collapse
|
23
|
Chen L, Ai F, Wu X, Yu W, Jin X, Ma J, Xiang B, Shen S, Li X. Analysis of neutrophil extracellular trap-related genes in Crohn's disease based on bioinformatics. J Cell Mol Med 2024; 28:e70013. [PMID: 39199011 PMCID: PMC11358036 DOI: 10.1111/jcmm.70013] [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: 02/18/2024] [Revised: 06/28/2024] [Accepted: 07/25/2024] [Indexed: 09/01/2024] Open
Abstract
Crohn's disease (CD) presents with diverse clinical phenotypes due to persistent inflammation of the gastrointestinal tract. Its global incidence is on the rise. Neutrophil extracellular traps (NETs) are networks released by neutrophils that capture microbicidal proteins and oxidases targeting pathogens. Research has shown that NETs are implicated in the pathogenesis of several immune-mediated diseases such as rheumatoid arthritis, systemic lupus erythematosus and inflammatory bowel disease. The goal of this study was to identify a panel of NET-related genes to construct a diagnostic and therapeutic model for CD. Through analysis of the GEO database, we identified 1950 differentially expressed genes (DEGs) associated with CD. Gene enrichment and immune cell infiltration analyses indicate that neutrophil infiltrates and chemokine-related pathways are predominantly involved in CD, with other immune cells such as CD4 and M1 macrophages also playing a role in disease progression. Utilizing weighted gene co-expression network analysis (WGCNA) and protein-protein interaction (PPI) networks, we identified six hub genes (SPP1, SOCS3, TIMP1, IRF1, CXCL2 and CD274). To validate the accuracy of our model, we performed external validation with statistical differences(p < 0.05). Additionally, immunohistochemical experiments demonstrated higher protein expression of the hub genes in colonic tissues from CD patients compared to healthy subjects (p < 0.05). In summary, we identified six effective hub genes associated with NETs as potential diagnostic markers for CD. These markers not only offer targets for future research but also hold promise for the development of novel therapeutic interventions for CD.
Collapse
Affiliation(s)
- Libin Chen
- Department of GastroenterologyThe Third Xiangya Hospital of Central South UniversityChangshaChina
- Hunan Key Laboratory of Nonresolving Inflammation and CancerThe Third Xiangya Hospital of Central South UniversityChangshaChina
| | - Feiyan Ai
- Department of GastroenterologyThe Third Xiangya Hospital of Central South UniversityChangshaChina
- Hunan Key Laboratory of Nonresolving Inflammation and CancerThe Third Xiangya Hospital of Central South UniversityChangshaChina
| | - Xing Wu
- Department of GastroenterologyThe Third Xiangya Hospital of Central South UniversityChangshaChina
- Hunan Key Laboratory of Nonresolving Inflammation and CancerThe Third Xiangya Hospital of Central South UniversityChangshaChina
| | - Wentao Yu
- Department of Pathology, The Third Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Xintong Jin
- Department of GastroenterologyThe Third Xiangya Hospital of Central South UniversityChangshaChina
- Hunan Key Laboratory of Nonresolving Inflammation and CancerThe Third Xiangya Hospital of Central South UniversityChangshaChina
| | - Jian Ma
- Hunan Key Laboratory of Nonresolving Inflammation and CancerThe Third Xiangya Hospital of Central South UniversityChangshaChina
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medical SciencesCentral South UniversityChangshaChina
| | - Bo Xiang
- Hunan Key Laboratory of Nonresolving Inflammation and CancerThe Third Xiangya Hospital of Central South UniversityChangshaChina
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medical SciencesCentral South UniversityChangshaChina
| | - Shourong Shen
- Department of GastroenterologyThe Third Xiangya Hospital of Central South UniversityChangshaChina
- Hunan Key Laboratory of Nonresolving Inflammation and CancerThe Third Xiangya Hospital of Central South UniversityChangshaChina
| | - Xiayu Li
- Department of GastroenterologyThe Third Xiangya Hospital of Central South UniversityChangshaChina
- Hunan Key Laboratory of Nonresolving Inflammation and CancerThe Third Xiangya Hospital of Central South UniversityChangshaChina
| |
Collapse
|
24
|
Yi K, An L, Qi Y, Yang T, Duan Y, Zhao X, Zhang P, Huang X, Su X, Tang Z, Sun D. Docosahexaenoic acid (DHA) promotes recovery from postoperative ileus and the repair of the injured intestinal barrier through mast cell-nerve crosstalk. Int Immunopharmacol 2024; 136:112316. [PMID: 38823183 DOI: 10.1016/j.intimp.2024.112316] [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: 02/11/2024] [Revised: 03/31/2024] [Accepted: 05/19/2024] [Indexed: 06/03/2024]
Abstract
The objective of this study was to investigate the neuroimmune mechanisms implicated in the enhancement of gastrointestinal function through the administration of oral DHA. Mast cell-deficient mice (KitW-sh) and C57BL/6 mice were used to establish postoperative ileus (POI) models. To further validate our findings, we conducted noncontact coculture experiments involving dorsal root ganglion (DRG) cells, bone marrow-derived mast cells (BMMCs) and T84 cells. Furthermore, the results obtained from investigations conducted on animals and cells were subsequently validated through clinical trials. The administration of oral DHA had ameliorative effects on intestinal barrier injury and postoperative ileus. In a mechanistic manner, the anti-inflammatory effect of DHA was achieved through the activation of transient receptor potential ankyrin 1 (TRPA1) on DRG cells, resulting in the stabilization of mast cells and increasing interleukin 10 (IL-10) secretion in mast cells. Furthermore, the activation of the pro-repair WNT1-inducible signaling protein 1 (WISP-1) signaling pathways by mast cell-derived IL-10 resulted in an enhancement of the intestinal barrier integrity. The current study demonstrated that the neuroimmune interaction between mast cells and nerves played a crucial role in the process of oral DHA improving the intestinal barrier integrity of POI, which further triggered the activation of CREB/WISP-1 signaling in intestinal mucosal cells.
Collapse
Affiliation(s)
- Keqian Yi
- Department of Gastrointestinal Surgery, Second Affiliated Hospital of Kunming Medical University/Second Faculty of Clinical Medicine, Kunming Medical University, Kunming 650101, China
| | - Liya An
- Department of Gastrointestinal Surgery, Second Affiliated Hospital of Kunming Medical University/Second Faculty of Clinical Medicine, Kunming Medical University, Kunming 650101, China
| | - Yuxing Qi
- Department of Gastrointestinal Surgery, Second Affiliated Hospital of Kunming Medical University/Second Faculty of Clinical Medicine, Kunming Medical University, Kunming 650101, China
| | - Ting Yang
- Department of Gastrointestinal Surgery, Second Affiliated Hospital of Kunming Medical University/Second Faculty of Clinical Medicine, Kunming Medical University, Kunming 650101, China
| | - Yongqing Duan
- Department of Gastrointestinal Surgery, Second Affiliated Hospital of Kunming Medical University/Second Faculty of Clinical Medicine, Kunming Medical University, Kunming 650101, China
| | - Xiaohu Zhao
- Department of Medicine, Monash University, Clayton, Victoria 3168, Australia
| | - Pengcheng Zhang
- Department of Gastrointestinal Surgery, Second Affiliated Hospital of Kunming Medical University/Second Faculty of Clinical Medicine, Kunming Medical University, Kunming 650101, China
| | - Xingzong Huang
- Department of Gastrointestinal Surgery, Second Affiliated Hospital of Kunming Medical University/Second Faculty of Clinical Medicine, Kunming Medical University, Kunming 650101, China
| | - Xianming Su
- Department of Gastrointestinal Surgery, Second Affiliated Hospital of Kunming Medical University/Second Faculty of Clinical Medicine, Kunming Medical University, Kunming 650101, China
| | - Zhiyi Tang
- Department of Gastroenterology, Second Affiliated Hospital of Kunming Medical University/Second Faculty of Clinical Medicine, Kunming Medical University, Kunming 650101, China.
| | - Dali Sun
- Department of Gastrointestinal Surgery, Second Affiliated Hospital of Kunming Medical University/Second Faculty of Clinical Medicine, Kunming Medical University, Kunming 650101, China.
| |
Collapse
|
25
|
Alam SB, Yan Z, Verma NH, Unsworth LD, Kulka M. Butyrate Increases Heparin Synthesis and Storage in Human Mast Cells. Cells 2024; 13:1241. [PMID: 39120272 PMCID: PMC11311861 DOI: 10.3390/cells13151241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 07/15/2024] [Accepted: 07/17/2024] [Indexed: 08/10/2024] Open
Abstract
Sulphated glycosaminoglycans (GAGs) such as heparin are a major component of mast cell granules and form the matrix within which biogenic mediators are stored. Since GAGs released from mast cells also play an important role in helminth expulsion, understanding GAG storage can offer new insights into mast cell function. Sodium butyrate (NaBu), a short-chain fatty acid, causes ultrastructural changes within the granules of human mast cells (HMC-1) and increases their histamine content. Therefore, we hypothesized that NaBu treatment would also modify the storage of polysaccharides such as GAGs. NaBu (1 mM) significantly increased GAG content and granularity in a time- and concentration-dependent manner without affecting cell viability and metabolic activity. NaBu increased the expression of enzymes associated with heparin biosynthesis (GLCE, NDST1, NDST2, HS6ST1, and GALT1) in a time-dependent manner. A cholesteryl butyrate emulsion (CholButE) increased heparin content after 24 and 48 h and modestly altered the expression of genes involved in heparin biosynthesis. Similar to NaBu, CholButE reduced cell proliferation without significantly altering viability or metabolic activity. These data show that butyrate increases the synthesis and storage of heparin in human mast cells, perhaps by altering their metabolic pathways.
Collapse
Affiliation(s)
- Syed Benazir Alam
- Quantum and Nanotechnologies Research Centre, National Research Council Canada, Edmonton, AB T6G 2M9, Canada; (S.B.A.); (Z.Y.); (N.H.V.)
| | - Zhimin Yan
- Quantum and Nanotechnologies Research Centre, National Research Council Canada, Edmonton, AB T6G 2M9, Canada; (S.B.A.); (Z.Y.); (N.H.V.)
| | - Nishita Hiresha Verma
- Quantum and Nanotechnologies Research Centre, National Research Council Canada, Edmonton, AB T6G 2M9, Canada; (S.B.A.); (Z.Y.); (N.H.V.)
- Department of Chemical and Materials Engineering, University of Alberta, Edmonton, AB T6G 1H9, Canada;
| | - Larry D. Unsworth
- Department of Chemical and Materials Engineering, University of Alberta, Edmonton, AB T6G 1H9, Canada;
- Department of Biomedical Engineering, University of Alberta, Edmonton, AB T6G 1H9, Canada
| | - Marianna Kulka
- Quantum and Nanotechnologies Research Centre, National Research Council Canada, Edmonton, AB T6G 2M9, Canada; (S.B.A.); (Z.Y.); (N.H.V.)
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB T6G 2E1, Canada
| |
Collapse
|
26
|
Ziaka M, Exadaktylos A. Gut-derived immune cells and the gut-lung axis in ARDS. Crit Care 2024; 28:220. [PMID: 38965622 PMCID: PMC11225303 DOI: 10.1186/s13054-024-05006-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Accepted: 06/26/2024] [Indexed: 07/06/2024] Open
Abstract
The gut serves as a vital immunological organ orchestrating immune responses and influencing distant mucosal sites, notably the respiratory mucosa. It is increasingly recognized as a central driver of critical illnesses, with intestinal hyperpermeability facilitating bacterial translocation, systemic inflammation, and organ damage. The "gut-lung" axis emerges as a pivotal pathway, where gut-derived injurious factors trigger acute lung injury (ALI) through the systemic circulation. Direct and indirect effects of gut microbiota significantly impact immune responses. Dysbiosis, particularly intestinal dysbiosis, termed as an imbalance of microbial species and a reduction in microbial diversity within certain bodily microbiomes, influences adaptive immune responses, including differentiating T regulatory cells (Tregs) and T helper 17 (Th17) cells, which are critical in various lung inflammatory conditions. Additionally, gut and bone marrow immune cells impact pulmonary immune activity, underscoring the complex gut-lung interplay. Moreover, lung microbiota alterations are implicated in diverse gut pathologies, affecting local and systemic immune landscapes. Notably, lung dysbiosis can reciprocally influence gut microbiota composition, indicating bidirectional gut-lung communication. In this review, we investigate the pathophysiology of ALI/acute respiratory distress syndrome (ARDS), elucidating the role of immune cells in the gut-lung axis based on recent experimental and clinical research. This exploration aims to enhance understanding of ALI/ARDS pathogenesis and to underscore the significance of gut-lung interactions in respiratory diseases.
Collapse
Affiliation(s)
- Mairi Ziaka
- Clinic of Geriatric Medicine, Center of Geriatric Medicine and Rehabilitation, Kantonsspital Baselland, Bruderholz, Switzerland.
- Department of Emergency Medicine, Inselspital, University Hospital, University of Bern, Bern, Switzerland.
| | - Aristomenis Exadaktylos
- Department of Emergency Medicine, Inselspital, University Hospital, University of Bern, Bern, Switzerland
| |
Collapse
|
27
|
Wei Z, Tang X, Yi C, Ocansey DKW, Mao F, Mao Z. HucMSC-Ex alleviates DSS-induced colitis in mice by decreasing mast cell activation via the IL-33/ST2 axis. Am J Transl Res 2024; 16:2727-2744. [PMID: 39006299 PMCID: PMC11236658 DOI: 10.62347/exze5413] [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: 05/22/2024] [Accepted: 06/24/2024] [Indexed: 07/16/2024]
Abstract
BACKGROUND Inflammatory bowel disease (IBD) is a chronic inflammatory disease that poses challenges in terms of treatment. The precise mechanism underlying the role of human umbilical cord mesenchymal stem cell-derived exosome (HucMSC-Ex) in the inflammatory repair process of IBD remains elusive. Mucosal mast cells accumulate within the intestinal tract and exert regulatory functions in IBD, thus presenting a novel target for addressing this intestinal disease. METHODS A mouse model of Dextran Sulfate Sodium (DSS)-induced colitis was established and hucMSC-Ex were administered to investigate their impact on the regulation of intestinal mast cells. An in vitro co-culture model using the human clonal colorectal adenocarcinoma cell line (Caco-2) and human mast cell line (LAD2) was also established for further exploration of the effect of hucMSC-Ex. RESULTS We observed the accumulation of mast cells in the intestines of patients with IBD as well as mice. In colitis mice, there was an upregulation of mast cell-related tryptase, interleukin-33 (IL-33), and suppression of tumorigenicity 2 receptor (ST2 or IL1RL1), and the function of the intestinal mucosal barrier related to intestinal tight junction protein was weakened. HucMSC-Ex treatment significantly reduced mast cell infiltration and intestinal damage. In the co-culture model, a substantial number of mast cells interact with the epithelial barrier, triggering activation of the IL-33/IL1RL1 (ST2) pathway and subsequent release of inflammatory factors and trypsin. This disruption leads to aberrant expression of tight junction proteins, which can be alleviated by supplementation with hucMSC-Ex. CONCLUSION Our results suggest that hucMSC-Ex may reduce the release of mast cell mediators via the IL-33/IL1RL1 (ST2) axis, thereby mitigating its detrimental effects on intestinal barrier function.
Collapse
Affiliation(s)
- Zhiping Wei
- Department of Laboratory Medicine, The Affiliated People’s Hospital, Jiangsu UniversityZhenjiang 212002, Jiangsu, P. R. China
- Department of Clinical Laboratory, The Third People’s Hospital of Xindu DistrictChengdu 610500, Sichuan, P. R. China
| | - Xiaohua Tang
- Department of Orthopaedics, The People’s Hospital of Danyang, Affiliated Danyang Hospital of Nantong UniversityZhenjiang 212300, Jiangsu, P. R. China
| | - Chengxue Yi
- School of Medical Technology, Zhenjiang CollegeZhenjiang 212028, Jiangsu, P. R. China
| | - Dickson Kofi Wiredu Ocansey
- Department of Laboratory Medicine, The Affiliated People’s Hospital, Jiangsu UniversityZhenjiang 212002, Jiangsu, P. R. China
- Department of Medical Laboratory Science, School of Allied Health Sciences, College of Health and Allied Sciences, University of Cape CoastCape Coast CC0959347, Ghana
| | - Fei Mao
- Department of Laboratory Medicine, The Affiliated People’s Hospital, Jiangsu UniversityZhenjiang 212002, Jiangsu, P. R. China
| | - Zhenwei Mao
- The Key Lab of Precision Diagnosis and Treatment in Hematologic Malignancies of Zhenjiang City, Affiliated People’s Hospital of Jiangsu UniversityZhenjiang 212002, Jiangsu, P. R. China
| |
Collapse
|
28
|
Billipp TE, Fung C, Webeck LM, Sargent DB, Gologorsky MB, Chen Z, McDaniel MM, Kasal DN, McGinty JW, Barrow KA, Rich LM, Barilli A, Sabat M, Debley JS, Wu C, Myers R, Howitt MR, von Moltke J. Tuft cell-derived acetylcholine promotes epithelial chloride secretion and intestinal helminth clearance. Immunity 2024; 57:1243-1259.e8. [PMID: 38744291 PMCID: PMC11168877 DOI: 10.1016/j.immuni.2024.03.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 02/05/2024] [Accepted: 03/29/2024] [Indexed: 05/16/2024]
Abstract
Epithelial cells secrete chloride to regulate water release at mucosal barriers, supporting both homeostatic hydration and the "weep" response that is critical for type 2 immune defense against parasitic worms (helminths). Epithelial tuft cells in the small intestine sense helminths and release cytokines and lipids to activate type 2 immune cells, but whether they regulate epithelial secretion is unknown. Here, we found that tuft cell activation rapidly induced epithelial chloride secretion in the small intestine. This response required tuft cell sensory functions and tuft cell-derived acetylcholine (ACh), which acted directly on neighboring epithelial cells to stimulate chloride secretion, independent of neurons. Maximal tuft cell-induced chloride secretion coincided with immune restriction of helminths, and clearance was delayed in mice lacking tuft cell-derived ACh, despite normal type 2 inflammation. Thus, we have uncovered an epithelium-intrinsic response unit that uses ACh to couple tuft cell sensing to the secretory defenses of neighboring epithelial cells.
Collapse
Affiliation(s)
- Tyler E Billipp
- Department of Immunology, University of Washington School of Medicine, Seattle, WA, USA
| | - Connie Fung
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Lily M Webeck
- Department of Immunology, University of Washington School of Medicine, Seattle, WA, USA
| | - Derek B Sargent
- Department of Immunology, University of Washington School of Medicine, Seattle, WA, USA
| | - Matthew B Gologorsky
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Zuojia Chen
- Experimental Immunology Branch, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Margaret M McDaniel
- Department of Immunology, University of Washington School of Medicine, Seattle, WA, USA
| | - Darshan N Kasal
- Department of Immunology, University of Washington School of Medicine, Seattle, WA, USA
| | - John W McGinty
- Department of Immunology, University of Washington School of Medicine, Seattle, WA, USA
| | - Kaitlyn A Barrow
- Center for Respiratory Biology and Therapeutics, Seattle Children's Research Institute, Seattle, WA, USA
| | - Lucille M Rich
- Center for Respiratory Biology and Therapeutics, Seattle Children's Research Institute, Seattle, WA, USA
| | | | - Mark Sabat
- Takeda Pharmaceuticals, San Diego, CA, USA
| | - Jason S Debley
- Center for Respiratory Biology and Therapeutics, Seattle Children's Research Institute, Seattle, WA, USA; Department of Pediatrics, Division of Pulmonary and Sleep Medicine, Seattle Children's Hospital, University of Washington, Seattle, WA, USA
| | - Chuan Wu
- Experimental Immunology Branch, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | | | - Michael R Howitt
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Jakob von Moltke
- Department of Immunology, University of Washington School of Medicine, Seattle, WA, USA.
| |
Collapse
|
29
|
Putro E, Carnevale A, Marangio C, Fulci V, Paolini R, Molfetta R. New Insight into Intestinal Mast Cells Revealed by Single-Cell RNA Sequencing. Int J Mol Sci 2024; 25:5594. [PMID: 38891782 PMCID: PMC11171657 DOI: 10.3390/ijms25115594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 05/14/2024] [Accepted: 05/18/2024] [Indexed: 06/21/2024] Open
Abstract
Mast cells (MCs) are tissue-resident immune cells distributed in all tissues and strategically located close to blood and lymphatic vessels and nerves. Thanks to the expression of a wide array of receptors, MCs act as tissue sentinels, able to detect the presence of bacteria and parasites and to respond to different environmental stimuli. MCs originate from bone marrow (BM) progenitors that enter the circulation and mature in peripheral organs under the influence of microenvironment factors, thus differentiating into heterogeneous tissue-specific subsets. Even though MC activation has been traditionally linked to IgE-mediated allergic reactions, a role for these cells in other pathological conditions including tumor progression has recently emerged. However, several aspects of MC biology remain to be clarified. The advent of single-cell RNA sequencing platforms has provided the opportunity to understand MCs' origin and differentiation as well as their phenotype and functions within different tissues, including the gut. This review recapitulates how single-cell transcriptomic studies provided insight into MC development as well as into the functional role of intestinal MC subsets in health and disease.
Collapse
Affiliation(s)
| | | | | | | | - Rossella Paolini
- Department of Molecular Medicine, Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University of Rome, 00161 Rome, Italy; (E.P.); (A.C.); (C.M.); (V.F.); (R.M.)
| | | |
Collapse
|
30
|
Wang Y, Tan Q, Pan M, Yu J, Wu S, Tu W, Li M, Jiang S. Minimally invasive vagus nerve stimulation modulates mast cell degranulation via the microbiota-gut-brain axis to ameliorate blood-brain barrier and intestinal barrier damage following ischemic stroke. Int Immunopharmacol 2024; 132:112030. [PMID: 38603861 DOI: 10.1016/j.intimp.2024.112030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 03/11/2024] [Accepted: 04/05/2024] [Indexed: 04/13/2024]
Abstract
Mast cells (MCs) play a significant role in various diseases, and their activation and degranulation can trigger inflammatory responses and barrier damage. Several studies have indicated that vagus nerve stimulation (VNS) exerts ameliorates neurological injury, and regulates gut MC degranulation. However, there is limited research on the modulatory effect of VNS on MCs in both the gut and brain in brain ischemia-reperfusion (I/R) injury in this process. We aim to develop a minimally invasive, targeted and convenient VNS approach to assess the impact of VNS and to clarify the relationship between VNS and MCs on the prognosis of acute ischemic stroke. We utilized middle cerebral artery occlusion/reperfusion (MCAO/r) to induce brain I/R injury. After the experiment, the motor function and neurofunctional impairments of the rats were detected, and the gastrointestinal function, blood-brain barrier (BBB) and intestinal barrier damage, and systemic and local inflammation were evaluated by Nissl, TTC staining, Evans blue, immunofluorescence staining, transmission electron microscopy, western blot assays, ELISA, and fecal 16S rRNA sequencing methods. Our research confirmed that our minimally invasive VNS method is a novel approach for stimulating the vagus nerve. VNS alleviated motor deficits and gastrointestinal dysfunction while also suppressing intestinal and neuroinflammation. Additionally, VNS ameliorated gut microbiota dysbiosis in rats. Furthermore, our analysis indicated that VNS reduces chymase secretion by modulating MCs degranulation and improves intestinal and BBB damage. Our results showed that VNS treatment can alleviate the damage of BBB and colonic barrier after cerebral I/R by modulating mast cell degranulation, and alleviates systemic inflammatory responses.
Collapse
Affiliation(s)
- Yanan Wang
- Rehabilitation Medicine Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Intelligent Rehabilitation Research Center, International Institute for Acupuncture and Rehabilitation, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; The Provincial Key Laboratory for Acupuncture and Rehabilitation in Zhejiang Province, The Wenzhou Key Laboratory for Rehabilitation Research, China
| | - Qianqian Tan
- Rehabilitation Medicine Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Intelligent Rehabilitation Research Center, International Institute for Acupuncture and Rehabilitation, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; The Provincial Key Laboratory for Acupuncture and Rehabilitation in Zhejiang Province, The Wenzhou Key Laboratory for Rehabilitation Research, China
| | - Mingdong Pan
- Rehabilitation Medicine Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Jiaying Yu
- Rehabilitation Medicine Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Intelligent Rehabilitation Research Center, International Institute for Acupuncture and Rehabilitation, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; The Provincial Key Laboratory for Acupuncture and Rehabilitation in Zhejiang Province, The Wenzhou Key Laboratory for Rehabilitation Research, China
| | - Shaoqi Wu
- Rehabilitation Medicine Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Intelligent Rehabilitation Research Center, International Institute for Acupuncture and Rehabilitation, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; The Provincial Key Laboratory for Acupuncture and Rehabilitation in Zhejiang Province, The Wenzhou Key Laboratory for Rehabilitation Research, China
| | - Wenzhan Tu
- Rehabilitation Medicine Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Intelligent Rehabilitation Research Center, International Institute for Acupuncture and Rehabilitation, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; The Provincial Key Laboratory for Acupuncture and Rehabilitation in Zhejiang Province, The Wenzhou Key Laboratory for Rehabilitation Research, China
| | - Ming Li
- School of Basic Medical Science, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China.
| | - Songhe Jiang
- Rehabilitation Medicine Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Intelligent Rehabilitation Research Center, International Institute for Acupuncture and Rehabilitation, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; The Provincial Key Laboratory for Acupuncture and Rehabilitation in Zhejiang Province, The Wenzhou Key Laboratory for Rehabilitation Research, China.
| |
Collapse
|
31
|
Chen Y, Feng S, Li Y, Zhang C, Chao G, Zhang S. Gut microbiota and intestinal immunity-A crosstalk in irritable bowel syndrome. Immunology 2024; 172:1-20. [PMID: 38174581 DOI: 10.1111/imm.13749] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 12/20/2023] [Indexed: 01/05/2024] Open
Abstract
Irritable bowel syndrome (IBS), one of the most prevalent functional gastrointestinal disorders, is characterized by recurrent abdominal pain and abnormal defecation habits, resulting in a severe healthcare burden worldwide. The pathophysiological mechanisms of IBS are multi-factorially involved, including food antigens, visceral hypersensitivity reactions, and the brain-gut axis. Numerous studies have found that gut microbiota and intestinal mucosal immunity play an important role in the development of IBS in crosstalk with multiple mechanisms. Therefore, based on existing evidence, this paper elaborates that the damage and activation of intestinal mucosal immunity and the disturbance of gut microbiota are closely related to the progression of IBS. Combined with the application prospect, it also provides references for further in-depth exploration and clinical practice.
Collapse
Affiliation(s)
- Yuxuan Chen
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Shuyan Feng
- The First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Ying Li
- The First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Chi Zhang
- Sir Run Run Shaw Hospital of Zhejiang University, Hangzhou, China
| | - Guanqun Chao
- Department of General Practice, Sir Run Run Shaw Hospital of Zhejiang University, Hangzhou, China
| | - Shuo Zhang
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
- Department of Gastroenterology, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
32
|
Cosoroaba R, Ceausu RA, Gaje NP, Vasca EM, Dumitru CS, Olariu I, Popovici RA, Raica M. High Intraepithelial Mast Cell Density in Warthin's Tumor. In Vivo 2024; 38:1104-1111. [PMID: 38688595 PMCID: PMC11059896 DOI: 10.21873/invivo.13544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 01/22/2024] [Accepted: 01/23/2024] [Indexed: 05/02/2024]
Abstract
BACKGROUND/AIM Warthin's tumor, the second most frequent neoplasia of the parotid gland, is characterized by a proliferation of both epithelial and lymphoid components. In addition to epithelial and lymphoid cells, various other cell types are implicated to varying degrees in the immune response. Notably, mast cells have long been recognized as a consistent cell population within this tumor. Despite the historical acknowledgment of mast cell presence, their true distribution and significance within Warthin's tumor remain unclear. In this study, we aimed to elucidate the distribution and significance of mast cells in Warthin's tumor. MATERIALS AND METHODS Histochemical and immunohistochemical methods were employed for the evaluation of mast cells within tumor specimens. RESULTS Our study revealed a notable concentration of mast cells in the epithelial component of Warthin's tumor. Microscopic examination showed predominant lymphoid and epithelial elements with occasional cystic formations. Immunohistochemical analysis identified mast cells in both components, emphasizing their role in the tumor microenvironment. Double immunostaining (mast cell tryptase and CD34) revealed no significant correlation between mast cells and blood vessels. Intraepithelial mast cells (IEMCs) had a significantly higher density in the epithelial component, suggesting a potential association with the tumor's benign nature. The relationship between IEMCs and epithelial cells, especially in the presence of cystic structures, offers valuable insights into the unique features of Warthin's tumor. CONCLUSION Our study contributes to the understanding of mast cells in Warthin's tumor, highlighting a substantial concentration within the epithelial component. This knowledge may pave the way for further investigations into the roles of mast cells in the pathogenesis and treatment of Warthin's tumor.
Collapse
Affiliation(s)
- Raluca Cosoroaba
- Department I, Faculty of Dental Medicine, Victor Babes University of Medicine and Pharmacy, Timisoara, Romania
| | - Raluca Amalia Ceausu
- Department of Microscopic Morphology/Histology, Angiogenesis Research Center, Victor Babes University of Medicine and Pharmacy, Timisoara, Romania
| | - Nela Pusa Gaje
- Department of Microscopic Morphology/Histology, Angiogenesis Research Center, Victor Babes University of Medicine and Pharmacy, Timisoara, Romania
| | - Elisabeta Maria Vasca
- Department of Dentistry, Faculty of Dentistry, Vasile Goldis Western University of Arad, Arad, Romania
| | - Cristina Stefania Dumitru
- Department of Microscopic Morphology/Histology, Angiogenesis Research Center, Victor Babes University of Medicine and Pharmacy, Timisoara, Romania;
| | - Iustin Olariu
- Department of Dentistry, Faculty of Dentistry, Vasile Goldis Western University of Arad, Arad, Romania
| | - Ramona Amina Popovici
- Department I, Faculty of Dental Medicine, Victor Babes University of Medicine and Pharmacy, Timisoara, Romania
| | - Marius Raica
- Department of Microscopic Morphology/Histology, Angiogenesis Research Center, Victor Babes University of Medicine and Pharmacy, Timisoara, Romania
| |
Collapse
|
33
|
Céspedes N, Fellows AM, Donnelly EL, Kaylor HL, Coles TA, Wild R, Dobson M, Schauer J, Van de Water J, Luckhart S. Basophil-Derived IL-4 and IL-13 Protect Intestinal Barrier Integrity and Control Bacterial Translocation during Malaria. Immunohorizons 2024; 8:371-383. [PMID: 38780542 PMCID: PMC11150129 DOI: 10.4049/immunohorizons.2300084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 04/29/2024] [Indexed: 05/25/2024] Open
Abstract
Our previous work demonstrated that basophils regulate a suite of malaria phenotypes, including intestinal mastocytosis and permeability, the immune response to infection, gametocytemia, and parasite transmission to the malaria mosquito Anopheles stephensi. Given that activated basophils are primary sources of the regulatory cytokines IL-4 and IL-13, we sought to examine the contributions of these mediators to basophil-dependent phenotypes in malaria. We generated mice with basophils depleted for IL-4 and IL-13 (baso IL-4/IL-13 (-)) and genotype controls (baso IL-4/IL-13 (+)) by crossing mcpt8-Cre and Il4/Il13fl/fl mice and infected them with Plasmodium yoelii yoelii 17XNL. Conditional deletion was associated with ileal mastocytosis and mast cell (MC) activation, increased intestinal permeability, and increased bacterial 16S levels in blood, but it had no effect on neutrophil activation, parasitemia, or transmission to A. stephensi. Increased intestinal permeability in baso IL-4/IL-13 (-) mice was correlated with elevated plasma eotaxin (CCL11), a potent eosinophil chemoattractant, and increased ileal MCs, proinflammatory IL-17A, and the chemokines MIP-1α (CCL3) and MIP-1β (CCL4). Blood bacterial 16S copies were positively but weakly correlated with plasma proinflammatory cytokines IFN-γ and IL-12p40, suggesting that baso IL-4/IL-13 (-) mice failed to control bacterial translocation into the blood during malaria infection. These observations suggest that basophil-derived IL-4 and IL-13 do not contribute to basophil-dependent regulation of parasite transmission, but these cytokines do orchestrate protection of intestinal barrier integrity after P. yoelii infection. Specifically, basophil-dependent IL-4/IL-13 control MC activation and prevent infection-induced intestinal barrier damage and bacteremia, perhaps via regulation of eosinophils, macrophages, and Th17-mediated inflammation.
Collapse
Affiliation(s)
- Nora Céspedes
- Department of Entomology, Plant Pathology and Nematology, University of Idaho, Moscow, ID
| | - Abigail M. Fellows
- Department of Entomology, Plant Pathology and Nematology, University of Idaho, Moscow, ID
| | | | - Hannah L. Kaylor
- Department of Entomology, Plant Pathology and Nematology, University of Idaho, Moscow, ID
| | - Taylor A. Coles
- Department of Entomology, Plant Pathology and Nematology, University of Idaho, Moscow, ID
| | - Ryan Wild
- Department of Entomology, Plant Pathology and Nematology, University of Idaho, Moscow, ID
| | - Megan Dobson
- Department of Entomology, Plant Pathology and Nematology, University of Idaho, Moscow, ID
| | - Joseph Schauer
- Division of Rheumatology, Allergy and Clinical Immunology, University of California, Davis, CA
| | - Judy Van de Water
- Division of Rheumatology, Allergy and Clinical Immunology, University of California, Davis, CA
| | - Shirley Luckhart
- Department of Biological Sciences, University of Idaho, Moscow, ID
| |
Collapse
|
34
|
Khoury P, Wechsler JB. Role of Mast Cells in Eosinophilic Gastrointestinal Diseases. Immunol Allergy Clin North Am 2024; 44:311-327. [PMID: 38575226 PMCID: PMC11220468 DOI: 10.1016/j.iac.2024.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/06/2024]
Abstract
Mast cells play a central role in the pathogenesis of eosinophilic gastrointestinal disorders (EGIDs), including eosinophilic esophagitis. Their interactions with immune and structural cells, involvement in tissue remodeling, and contribution to symptoms make them attractive targets for therapeutic intervention. More is being discovered regarding the intricate interplay of mast cells and eosinophils. Recent studies demonstrating that depletion of eosinophils is insufficient to improve symptoms of EGIDs have raised the question of whether other cells may play a role in symptomatology and pathogenesis of EGIDs.
Collapse
Affiliation(s)
- Paneez Khoury
- Human Eosinophil Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 9000 Rockville Pike, Building 10, Room 12C103, Bethesda, MD 20892, USA.
| | - Joshua B Wechsler
- Simpson-Querrey 10-518, Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, 225 E. Chicago Avenue, Box 65, Chicago, IL 60611, USA
| |
Collapse
|
35
|
Bao C, Abraham SN. Mast cell-sensory neuron crosstalk in allergic diseases. J Allergy Clin Immunol 2024; 153:939-953. [PMID: 38373476 PMCID: PMC10999357 DOI: 10.1016/j.jaci.2024.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 01/12/2024] [Accepted: 02/14/2024] [Indexed: 02/21/2024]
Abstract
Mast cells (MCs) are tissue-resident immune cells, well-positioned at the host-environment interface for detecting external antigens and playing a critical role in mobilizing innate and adaptive immune responses. Sensory neurons are afferent neurons innervating most areas of the body but especially in the periphery, where they sense external and internal signals and relay information to the brain. The significance of MC-sensory neuron communication is now increasingly becoming recognized, especially because both cell types are in close physical proximity at the host-environment interface and around major organs of the body and produce specific mediators that can activate each other. In this review, we explore the roles of MC-sensory neuron crosstalk in allergic diseases, shedding light on how activated MCs trigger sensory neurons to initiate signaling in pruritus, shock, and potentially abdominal pain in allergy, and how activated sensory neurons regulate MCs in homeostasis and atopic dermatitis associated with contact hypersensitivity and type 2 inflammation. Throughout the review, we also discuss how these 2 sentinel cell types signal each other, potentially resulting in a positive feedback loop that can sustain inflammation. Unraveling the mysteries of MC-sensory neuron crosstalk is likely to unveil their critical roles in various disease conditions and enable the development of new therapeutic approaches to combat these maladies.
Collapse
Affiliation(s)
- Chunjing Bao
- Department of Pathology, Duke University Medical Center, Durham, NC
| | - Soman N Abraham
- Department of Pathology, Duke University Medical Center, Durham, NC; Department of Immunology, Duke University Medical Center, Durham, NC; Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC; Department of Program in Emerging Infectious Diseases, Duke-National University of Singapore, Singapore, Singapore.
| |
Collapse
|
36
|
Zhang DW, Lu JL, Dong BY, Fang MY, Xiong X, Qin XJ, Fan XM. Gut microbiota and its metabolic products in acute respiratory distress syndrome. Front Immunol 2024; 15:1330021. [PMID: 38433840 PMCID: PMC10904571 DOI: 10.3389/fimmu.2024.1330021] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 01/30/2024] [Indexed: 03/05/2024] Open
Abstract
The prevalence rate of acute respiratory distress syndrome (ARDS) is estimated at approximately 10% in critically ill patients worldwide, with the mortality rate ranging from 17% to 39%. Currently, ARDS mortality is usually higher in patients with COVID-19, giving another challenge for ARDS treatment. However, the treatment efficacy for ARDS is far from satisfactory. The relationship between the gut microbiota and ARDS has been substantiated by relevant scientific studies. ARDS not only changes the distribution of gut microbiota, but also influences intestinal mucosal barrier through the alteration of gut microbiota. The modulation of gut microbiota can impact the onset and progression of ARDS by triggering dysfunctions in inflammatory response and immune cells, oxidative stress, cell apoptosis, autophagy, pyroptosis, and ferroptosis mechanisms. Meanwhile, ARDS may also influence the distribution of metabolic products of gut microbiota. In this review, we focus on the impact of ARDS on gut microbiota and how the alteration of gut microbiota further influences the immune function, cellular functions and related signaling pathways during ARDS. The roles of gut microbiota-derived metabolites in the development and occurrence of ARDS are also discussed.
Collapse
Affiliation(s)
- Dong-Wei Zhang
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Inflammation & Allergic Diseases Research Unit, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Department of Respiratory and Critical Care Medicine, Liuzhou People’s Hospital, Guangxi Medical University, Liuzhou, Guangxi, China
- Key Laboratory of Diagnosis, Treatment and Research of Asthma and Chronic Obstructive Pulmonary Disease, Liuzhou, Guangxi, China
| | - Jia-Li Lu
- Department of Respiratory and Critical Care Medicine, Liuzhou People’s Hospital, Guangxi Medical University, Liuzhou, Guangxi, China
- Key Laboratory of Diagnosis, Treatment and Research of Asthma and Chronic Obstructive Pulmonary Disease, Liuzhou, Guangxi, China
| | - Bi-Ying Dong
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Inflammation & Allergic Diseases Research Unit, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Department of Respiratory and Critical Care Medicine, Liuzhou People’s Hospital, Guangxi Medical University, Liuzhou, Guangxi, China
- Key Laboratory of Diagnosis, Treatment and Research of Asthma and Chronic Obstructive Pulmonary Disease, Liuzhou, Guangxi, China
| | - Meng-Ying Fang
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Inflammation & Allergic Diseases Research Unit, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Xia Xiong
- Department of Dermatology, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Xue-Jun Qin
- Department of Respiratory and Critical Care Medicine, Liuzhou People’s Hospital, Guangxi Medical University, Liuzhou, Guangxi, China
- Key Laboratory of Diagnosis, Treatment and Research of Asthma and Chronic Obstructive Pulmonary Disease, Liuzhou, Guangxi, China
| | - Xian-Ming Fan
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Inflammation & Allergic Diseases Research Unit, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
37
|
Kvidera SK, Mayorga EJ, McCarthy CS, Horst EA, Abeyta MA, Baumgard LH. Effects of supplemental citrulline on thermal and intestinal morphology parameters during heat stress and feed restriction in growing pigs. J Anim Sci 2024; 102:skae120. [PMID: 38812469 PMCID: PMC11143481 DOI: 10.1093/jas/skae120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 05/29/2024] [Indexed: 05/31/2024] Open
Abstract
Study objectives were to characterize the effects of citrulline (CIT) on physiological and intestinal morphology metrics during heat stress (HS) and feed restriction. Forty crossbred gilts (30 ± 2 kg body weight [BW]) were assigned to one of five treatments: (1) thermoneutral (TN) fed ad libitum (AL) with control (CON) supplement (TNAL; n = 8), (2) TN pair-fed (PF) with CON (PF-CON; n = 8), (3) TN PF with CIT (PF-CIT; n = 8), (4) HS AL with CON (HS-CON; n = 8), and (5) HS AL with CIT (HS-CIT; n = 8). During the period (P) 1 (7 d), pigs were in TN conditions (23.6 °C) and fed AL their respective supplemental treatments. During P2 (2.5 d), HS-CON and HS-CIT pigs were fed AL and exposed to cyclical HS (33.6 to 38.3 °C), while TNAL, PF-CON, and PF-CIT remained in TN and were fed either AL or PF to their HS counterparts. Citrulline (0.13 g/kg BW) was orally administered twice daily during P1 and P2. HS increased rectal temperature (Tr), skin temperature (Ts), and respiration rate (RR) relative to TN pigs (0.8 °C, 4.7 °C, and 47 breaths/min, respectively; P < 0.01). However, HS-CIT had decreased RR (7 breaths/min, P = 0.04) and a tendency for decreased Tr (0.1 °C, P = 0.07) relative to HS-CON pigs. During P2, HS pigs had decreased feed intake (22%; P < 0.01) and a tendency for decreased average daily gain (P = 0.08) relative to TNAL pigs, and by experimental design, PF pigs followed this same pattern. Circulating lipopolysaccharide-binding protein tended to be decreased (29%; P = 0.08) in PF relative to TNAL pigs and was increased (41%; P = 0.03) in HS compared to PF pigs. Jejunum villus height was decreased in PF relative to TNAL pigs (15%; P = 0.03); however, CIT supplementation improved this metric during feed restriction (16%; P = 0.10). Jejunum mucosal surface area decreased in PF (16%; P = 0.02) and tended to decrease in HS (11%; P = 0.10) compared to TNAL pigs. Ileum villus height and mucosal surface area decreased in HS compared to TNAL pigs (10 and 14%, respectively; P ≤ 0.04), but both parameters were rescued by CIT supplementation (P ≤ 0.08). Intestinal myeloperoxidase and goblet cell area remained similar among treatments and intestinal segments (P > 0.24). In summary, CIT supplementation slightly improved RR and Tr during HS. Feed restriction and HS differentially affected jejunum and ileum morphology and while CIT ameliorated some of these effects, the benefit appeared dependent on intestinal section and stressor type.
Collapse
Affiliation(s)
- Sara K Kvidera
- Department of Animal Science, Iowa State University, Ames, Iowa 50011, USA
| | - Edith J Mayorga
- Department of Animal Science, Iowa State University, Ames, Iowa 50011, USA
| | - Carrie S McCarthy
- Department of Animal Science, Iowa State University, Ames, Iowa 50011, USA
| | - Erin A Horst
- Department of Animal Science, Iowa State University, Ames, Iowa 50011, USA
| | - Megan A Abeyta
- Department of Animal Science, Iowa State University, Ames, Iowa 50011, USA
| | - Lance H Baumgard
- Department of Animal Science, Iowa State University, Ames, Iowa 50011, USA
| |
Collapse
|
38
|
Mayorga EJ, Rodriguez-Jimenez S, Abeyta MA, Goetz BM, Opgenorth J, Moeser AJ, Baumgard LH. Investigating intestinal mast cell dynamics during acute heat stress in growing pigs. J Anim Sci 2024; 102:skae030. [PMID: 38290531 PMCID: PMC10889722 DOI: 10.1093/jas/skae030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 01/26/2024] [Indexed: 02/01/2024] Open
Abstract
Objectives were to examine the temporal pattern of intestinal mast cell dynamics and the effects of a mast cell stabilizer (ketotifen [Ket]) during acute heat stress (HS) in growing pigs. Crossbred barrows (n = 42; 32.3 ± 1.9 kg body weight [BW]) were randomly assigned to 1 of 7 environmental-therapeutic treatments: (1) thermoneutral (TN) control (TNCon; n = 6), (2) 2 h HS control (2 h HSCon; n = 6), (3) 2 h HS + Ket (2 h HSKet; n = 6); (4) 6 h HSCon (n = 6), (5) 6 h HSKet (n = 6), (6) 12 h HSCon (n = 6), or (7) 12 h HSKet (n = 6). Following 5 d of acclimation to individual pens, pigs were enrolled in two experimental periods (P). During P1 (3 d), pigs were housed in TN conditions (21.5 ± 0.8 °C) for the collection of baseline measurements. During P2, TNCon pigs remained in TN conditions for 12 h, while HS pigs were exposed to constant HS (38.1 ± 0.2 °C) for either 2, 6, or 12 h. Pigs were euthanized at the end of P2, and blood and tissue samples were collected. Regardless of time or therapeutic treatment, pigs exposed to HS had increased rectal temperature, skin temperature, and respiration rate compared to their TNCon counterparts (1.9 °C, 6.9° C, and 119 breaths/min; P < 0.01). As expected, feed intake and BW gain markedly decreased in HS pigs relative to their TNCon counterparts (P < 0.01). Irrespective of therapeutic treatment, circulating corticotropin-releasing factor decreased from 2 to 12 h of HS relative to TNCon pigs (P < 0.01). Blood cortisol increased at 2 h of HS (2-fold; P = 0.04) and returned to baseline by 6 h. Plasma histamine (a proxy of mast cell activation) remained similar across thermal treatments and was not affected by Ket administration (P > 0.54). Independent of Ket or time, HS increased mast cell numbers in the jejunum (94%; P < 0.01); however, no effects of HS on mast cell numbers were detected in the ileum or colon. Jejunum and ileum myeloperoxidase area remained similar among treatments (P > 0.58) but it tended to increase (12%; P = 0.08) in the colon in HSCon relative to TNCon pigs. Circulating lymphocytes and basophils decreased in HSKet relative to TN and HSCon pigs (P ≤ 0.06). Blood monocytes and eosinophils were reduced in HS pigs relative to their TNCon counterparts (P < 0.01). In summary, HS increased jejunum mast cell numbers and altered leukocyte dynamics and proinflammatory biomarkers. However, Ket administration had no effects on mast cell dynamics measured herein.
Collapse
Affiliation(s)
- Edith J Mayorga
- Department of Animal Science, Iowa State University, Ames, IA 50011, USA
| | | | - Megan A Abeyta
- Department of Animal Science, Iowa State University, Ames, IA 50011, USA
| | - Brady M Goetz
- Department of Animal Science, Iowa State University, Ames, IA 50011, USA
| | - Julie Opgenorth
- Department of Animal Science, Iowa State University, Ames, IA 50011, USA
| | - Adam J Moeser
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine, Michigan State University, East Lansing, MI 48824, USA
| | - Lance H Baumgard
- Department of Animal Science, Iowa State University, Ames, IA 50011, USA
| |
Collapse
|
39
|
Vicente-Santos A, Lock LR, Allira M, Dyer KE, Dunsmore A, Tu W, Volokhov DV, Herrera C, Lei GS, Relich RF, Janech MG, Bland AM, Simmons NB, Becker DJ. Serum proteomics reveals a tolerant immune phenotype across multiple pathogen taxa in wild vampire bats. Front Immunol 2023; 14:1281732. [PMID: 38193073 PMCID: PMC10773587 DOI: 10.3389/fimmu.2023.1281732] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 11/24/2023] [Indexed: 01/10/2024] Open
Abstract
Bats carry many zoonotic pathogens without showing pronounced pathology, with a few exceptions. The underlying immune tolerance mechanisms in bats remain poorly understood, although information-rich omics tools hold promise for identifying a wide range of immune markers and their relationship with infection. To evaluate the generality of immune responses to infection, we assessed the differences and similarities in serum proteomes of wild vampire bats (Desmodus rotundus) across infection status with five taxonomically distinct pathogens: bacteria (Bartonella spp., hemoplasmas), protozoa (Trypanosoma cruzi), and DNA (herpesviruses) and RNA (alphacoronaviruses) viruses. From 19 bats sampled in 2019 in Belize, we evaluated the up- and downregulated immune responses of infected versus uninfected individuals for each pathogen. Using a high-quality genome annotation for vampire bats, we identified 586 serum proteins but found no evidence for differential abundance nor differences in composition between infected and uninfected bats. However, using receiver operating characteristic curves, we identified four to 48 candidate biomarkers of infection depending on the pathogen, including seven overlapping biomarkers (DSG2, PCBP1, MGAM, APOA4, DPEP1, GOT1, and IGFALS). Enrichment analysis of these proteins revealed that our viral pathogens, but not the bacteria or protozoa studied, were associated with upregulation of extracellular and cytoplasmatic secretory vesicles (indicative of viral replication) and downregulation of complement activation and coagulation cascades. Additionally, herpesvirus infection elicited a downregulation of leukocyte-mediated immunity and defense response but an upregulation of an inflammatory and humoral immune response. In contrast to our two viral infections, we found downregulation of lipid and cholesterol homeostasis and metabolism with Bartonella spp. infection, of platelet-dense and secretory granules with hemoplasma infection, and of blood coagulation pathways with T. cruzi infection. Despite the small sample size, our results suggest that vampire bats have a similar suite of immune mechanisms for viruses distinct from responses to the other pathogen taxa, and we identify potential biomarkers that can expand our understanding of pathogenesis of these infections in bats. By applying a proteomic approach to a multi-pathogen system in wild animals, our study provides a distinct framework that could be expanded across bat species to increase our understanding of how bats tolerate pathogens.
Collapse
Affiliation(s)
| | - Lauren R. Lock
- School of Biological Sciences, University of Oklahoma, Norman, OK, United States
| | - Meagan Allira
- School of Biological Sciences, University of Oklahoma, Norman, OK, United States
| | - Kristin E. Dyer
- School of Biological Sciences, University of Oklahoma, Norman, OK, United States
| | - Annalise Dunsmore
- Department of Tropical Medicine, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, United States
- Vector-Borne and Infectious Disease Research Center, Tulane University, New Orleans, LA, United States
| | - Weihong Tu
- Department of Tropical Medicine, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, United States
- Vector-Borne and Infectious Disease Research Center, Tulane University, New Orleans, LA, United States
| | - Dmitriy V. Volokhov
- Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, United States
| | - Claudia Herrera
- Department of Tropical Medicine, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, United States
- Vector-Borne and Infectious Disease Research Center, Tulane University, New Orleans, LA, United States
| | - Guang-Sheng Lei
- Department of Pathology and Laboratory Medicine, School of Medicine, Indiana University, Indianapolis, IN, United States
| | - Ryan F. Relich
- Department of Pathology and Laboratory Medicine, School of Medicine, Indiana University, Indianapolis, IN, United States
| | - Michael G. Janech
- Hollings Marine Laboratory, Charleston, SC, United States
- Department of Biology, College of Charleston, Charleston, SC, United States
| | - Alison M. Bland
- Hollings Marine Laboratory, Charleston, SC, United States
- Department of Biology, College of Charleston, Charleston, SC, United States
| | - Nancy B. Simmons
- Department of Mammalogy, Division of Vertebrate Zoology, American Museum of Natural History, New York, NY, United States
| | - Daniel J. Becker
- School of Biological Sciences, University of Oklahoma, Norman, OK, United States
| |
Collapse
|
40
|
La Torre D, Van Oudenhove L, Vanuytsel T, Verbeke K. Psychosocial stress-induced intestinal permeability in healthy humans: What is the evidence? Neurobiol Stress 2023; 27:100579. [PMID: 37842017 PMCID: PMC10569989 DOI: 10.1016/j.ynstr.2023.100579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 10/04/2023] [Accepted: 10/05/2023] [Indexed: 10/17/2023] Open
Abstract
An impaired intestinal barrier function can be detrimental to the host as it may allow the translocation of luminal antigens and toxins into the subepithelial tissue and bloodstream. In turn, this may cause local and systemic immune responses and lead to the development of pathologies. In vitro and animal studies strongly suggest that psychosocial stress is one of the factors that can increase intestinal permeability via mast-cell dependent mechanisms. Remarkably, studies have not been able to yield unequivocal evidence that such relation between stress and intestinal permeability also exists in (healthy) humans. In the current Review, we discuss the mechanisms that are involved in stress-induced intestinal permeability changes and postulate factors that influence these alterations and that may explain the translational difficulties from in vitro and animal to human studies. As human research differs highly from animal research in the extent to which stress can be applied and intestinal permeability can be measured, it remains difficult to draw conclusions about the presence of a relation between stress and intestinal permeability in (healthy) humans. Future studies should bear in mind these difficulties, and more research into in vivo methods to assess intestinal permeability are warranted.
Collapse
Affiliation(s)
- Danique La Torre
- Translational Research Center in Gastrointestinal Disorders (TARGID), Department of Chronic Diseases and Metabolism, Faculty of Medicine, KU Leuven, Leuven, Belgium
- Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Lukas Van Oudenhove
- Translational Research Center in Gastrointestinal Disorders (TARGID), Department of Chronic Diseases and Metabolism, Faculty of Medicine, KU Leuven, Leuven, Belgium
- Leuven Brain Institute, KU Leuven, Leuven, Belgium
- Cognitive and Affective Neuroscience Lab, Department of Psychological and Brain Sciences, Dartmouth College, Hanover, NH, USA
| | - Tim Vanuytsel
- Translational Research Center in Gastrointestinal Disorders (TARGID), Department of Chronic Diseases and Metabolism, Faculty of Medicine, KU Leuven, Leuven, Belgium
- Division of Gastroenterology and Hepatology, Leuven University Hospital, Leuven, Belgium
| | - Kristin Verbeke
- Translational Research Center in Gastrointestinal Disorders (TARGID), Department of Chronic Diseases and Metabolism, Faculty of Medicine, KU Leuven, Leuven, Belgium
| |
Collapse
|
41
|
Rodrigues VMM, Bitencourt KCQM, Schapochnik A, de Souza V, da Palma Cruz M, Damazo AS, Ferreira CM, Cecatto RB, Hamblin MR, Destro Rodrigues MFS, Lino-Dos-Santos-Franco A. Comparison between local abdominal and transcutaneous tail vein photobiomodulation in experimental rat model of ulcerative colitis. Lasers Med Sci 2023; 38:247. [PMID: 37897531 DOI: 10.1007/s10103-023-03910-2] [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: 08/31/2023] [Accepted: 10/14/2023] [Indexed: 10/30/2023]
Abstract
Ulcerative colitis (UC) is a chronic autoimmune disease that impacts the quality of life, but current pharmacological treatments are limited. Photobiomodulation (PBM) is a light-based treatment that can be applied either locally or systemically. Here, we compare the effects of local and vascular PBM (VPBM) in an experimental rat model of UC. Male Wistar rats were induced with UC by rectal instillation of acetic acid and treated with either local abdominal PBM or VPBM to the tail vein using a 660-nm LED. The findings indicated that local PBM but not VPBM reduced intestinal histological scores. Both local and VPBM increased mucus production, decreased mast cell degranulation, and modulated TNF-α and IL-1 β levels in the intestines. Local PBM also affected the expression of the mRNAs for IL-6, TNF-α, and IFN-γ. In conclusion, we suggest that local PBM appears to be more promising than VPBM for treating UC. However, further research is needed to fully understand the mechanisms and to optimize the parameters of PBM for UC treatment.
Collapse
Affiliation(s)
- Virgínia Mendes Matias Rodrigues
- Post Graduate Program in Biophotonics Medicine, University Nove de Julho (UNINOVE), Rua Vergueiro, 239/245, São Paulo, SP, 01504-000, Brazil
| | | | - Adriana Schapochnik
- Post Graduate Program in Biophotonics Medicine, University Nove de Julho (UNINOVE), Rua Vergueiro, 239/245, São Paulo, SP, 01504-000, Brazil
| | - Vanessa de Souza
- Post Graduate Program in Biophotonics Medicine, University Nove de Julho (UNINOVE), Rua Vergueiro, 239/245, São Paulo, SP, 01504-000, Brazil
| | - Marlon da Palma Cruz
- Post Graduate Program in Biophotonics Medicine, University Nove de Julho (UNINOVE), Rua Vergueiro, 239/245, São Paulo, SP, 01504-000, Brazil
| | - Amílcar Sabino Damazo
- Department of Basic Science in Health, Faculty of Medical Sciences, Federal University of Cuiabá, Cuiabá, Brazil
| | | | - Rebeca Boltes Cecatto
- Post Graduate Program in Biophotonics Medicine, University Nove de Julho (UNINOVE), Rua Vergueiro, 239/245, São Paulo, SP, 01504-000, Brazil
| | - Michael R Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein, 2028, South Africa
| | | | - Adriana Lino-Dos-Santos-Franco
- Post Graduate Program in Biophotonics Medicine, University Nove de Julho (UNINOVE), Rua Vergueiro, 239/245, São Paulo, SP, 01504-000, Brazil.
| |
Collapse
|
42
|
Miranda Júnior NRD, Santos AGAD, Pereira AV, Mariano IA, Guilherme ALF, Santana PDL, Beletini LDF, Evangelista FF, Nogueira-Melo GDA, Sant'Ana DDMG. Rosuvastatin enhances alterations caused by Toxoplasma gondii in the duodenum of mice. Tissue Cell 2023; 84:102194. [PMID: 37597359 DOI: 10.1016/j.tice.2023.102194] [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] [Revised: 08/04/2023] [Accepted: 08/07/2023] [Indexed: 08/21/2023]
Abstract
Infection by Toxoplasma gondii may compromise the intestinal histoarchitecture through the tissue reaction triggered by the parasite. Thus, this study evaluated whether treatment with rosuvastatin modifies duodenal changes caused by the chronic infection induced by cysts of T. gondii. For this, female Swiss mice were distributed into infected and treated group (ITG), infected group (IG), group treated with 40 mg/kg rosuvastatin (TG) and control group (CG). After 72 days of infection, the animals were euthanized, the duodenum was collected and processed for histopathological analysis. We observed an increase in immune cell infiltration in the IG, TG and ITG groups, with injury to the Brunner glands. The infection led to a reduction in collagen fibers and mast cells. Infected and treated animals showed an increase in collagen fibers, acidic mucin-producing goblet cells, intraepithelial lymphocytes and mast cells, in addition to the reduction of muscle, neutral mucin-producing and Paneth cells. While treatment with rosuvastatin alone led to increased muscle layer, proportion of neutral mucin-producing goblet cells, Paneth cells, and reduction of collagen fibers. These findings indicate that the infection and treatment caused changes in the homeostasis of the intestinal wall and treatment with rosuvastatin potentiated most parameters indicative of inflammation.
Collapse
Affiliation(s)
- Nelson Raimundo de Miranda Júnior
- Biosciences and Physiopathology Program, State University of Maringá, Maringá, Brazil; Department of Basic Health Sciences, State University of Maringá, Maringá, Brazil
| | - Amanda Gubert Alves Dos Santos
- Biosciences and Physiopathology Program, State University of Maringá, Maringá, Brazil; Department of Basic Health Sciences, State University of Maringá, Maringá, Brazil
| | - Andréia Vieira Pereira
- Biosciences and Physiopathology Program, State University of Maringá, Maringá, Brazil; Department of Basic Health Sciences, State University of Maringá, Maringá, Brazil
| | - Isabela Alessandra Mariano
- Department of Clinical Analysis and Biomedicine, State University of Maringá, Maringá, Brazil; Department of Basic Health Sciences, State University of Maringá, Maringá, Brazil
| | - Ana Lucia Falavigna Guilherme
- Health Sciences Program, State University of Maringá, Maringá, Brazil; Department of Basic Health Sciences, State University of Maringá, Maringá, Brazil
| | - Priscilla de Laet Santana
- Health Sciences Program, State University of Maringá, Maringá, Brazil; Department of Basic Health Sciences, State University of Maringá, Maringá, Brazil
| | - Lucimara de Fátima Beletini
- Health Sciences Program, State University of Maringá, Maringá, Brazil; Department of Basic Health Sciences, State University of Maringá, Maringá, Brazil
| | - Fernanda Ferreira Evangelista
- Health Sciences Program, State University of Maringá, Maringá, Brazil; Department of Basic Health Sciences, State University of Maringá, Maringá, Brazil
| | - Gessilda de Alcantara Nogueira-Melo
- Biosciences and Physiopathology Program, State University of Maringá, Maringá, Brazil; Department of Basic Health Sciences, State University of Maringá, Maringá, Brazil
| | - Debora de Mello Gonçales Sant'Ana
- Biosciences and Physiopathology Program, State University of Maringá, Maringá, Brazil; Department of Basic Health Sciences, State University of Maringá, Maringá, Brazil.
| |
Collapse
|
43
|
Leigh SJ, Uhlig F, Wilmes L, Sanchez-Diaz P, Gheorghe CE, Goodson MS, Kelley-Loughnane N, Hyland NP, Cryan JF, Clarke G. The impact of acute and chronic stress on gastrointestinal physiology and function: a microbiota-gut-brain axis perspective. J Physiol 2023; 601:4491-4538. [PMID: 37756251 DOI: 10.1113/jp281951] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 09/06/2023] [Indexed: 09/29/2023] Open
Abstract
The physiological consequences of stress often manifest in the gastrointestinal tract. Traumatic or chronic stress is associated with widespread maladaptive changes throughout the gut, although comparatively little is known about the effects of acute stress. Furthermore, these stress-induced changes in the gut may increase susceptibility to gastrointestinal disorders and infection, and impact critical features of the neural and behavioural consequences of the stress response by impairing gut-brain axis communication. Understanding the mechanisms behind changes in enteric nervous system circuitry, visceral sensitivity, gut barrier function, permeability, and the gut microbiota following stress is an important research objective with pathophysiological implications in both neurogastroenterology and psychiatry. Moreover, the gut microbiota has emerged as a key aspect of physiology sensitive to the effects of stress. In this review, we focus on different aspects of the gastrointestinal tract including gut barrier function as well as the immune, humoral and neuronal elements involved in gut-brain communication. Furthermore, we discuss the evidence for a role of stress in gastrointestinal disorders. Existing gaps in the current literature are highlighted, and possible avenues for future research with an integrated physiological perspective have been suggested. A more complete understanding of the spatial and temporal dynamics of the integrated host and microbial response to different kinds of stressors in the gastrointestinal tract will enable full exploitation of the diagnostic and therapeutic potential in the fast-evolving field of host-microbiome interactions.
Collapse
Affiliation(s)
- Sarah-Jane Leigh
- APC Microbiome Ireland, Cork, Ireland
- Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland
| | - Friederike Uhlig
- APC Microbiome Ireland, Cork, Ireland
- Department of Physiology, University College Cork, Cork, Ireland
| | - Lars Wilmes
- APC Microbiome Ireland, Cork, Ireland
- Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Paula Sanchez-Diaz
- APC Microbiome Ireland, Cork, Ireland
- Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland
| | - Cassandra E Gheorghe
- APC Microbiome Ireland, Cork, Ireland
- Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland
| | - Michael S Goodson
- 711th Human Performance Wing, Air Force Research Laboratory, Wright-Patterson Air Force Base, Dayton, Ohio, USA
| | - Nancy Kelley-Loughnane
- Materials and Manufacturing Directorate, Air Force Research Laboratory, Wright-Patterson Air Force Base, Dayton, Ohio, USA
| | - Niall P Hyland
- APC Microbiome Ireland, Cork, Ireland
- Department of Physiology, University College Cork, Cork, Ireland
| | - John F Cryan
- APC Microbiome Ireland, Cork, Ireland
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Gerard Clarke
- APC Microbiome Ireland, Cork, Ireland
- Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland
| |
Collapse
|
44
|
Thongrin T, Suyapoh W, Wendo W, Tangkawattana P, Sukon P, Salao K, Suttiprapa S, Saichua P, Tangkawatana S. Inflammatory cell responses in biliary mucosa during Opisthorchis viverrini infection: Insights into susceptibility differences among hosts. Open Vet J 2023; 13:1150-1166. [PMID: 37842106 PMCID: PMC10576576 DOI: 10.5455/ovj.2023.v13.i9.11] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 08/15/2023] [Indexed: 10/17/2023] Open
Abstract
Background Individual host susceptibility is believed to be a risk factor in the interaction between the host and the parasite. Since studying time series in humans is limited, animal models are replaced. Aim This study aims to explore and compare the pattern of inflammatory cell types along the biliary tract and their association with proliferative lesions in the early development of cholangiocarcinoma from susceptible and nonsusceptible animal models. Methods Thirty male Syrian golden hamsters and 30 BALB/c mice, serving as the susceptible and nonsusceptible animal models, were used in this comparative study. The animals were infected with 50 Opisthorchis viverrini metacercariae via gastric intubation. At days 1, 2, 7, 14, 28, and 56 postinfection (p.i.), five animals were randomly selected from each group and humanely sacrificed. The hepatobiliary tissues were collected and processed for histopathological study. Histochemical and immunohistochemical staining were applied to differentiate the inflammatory cell types. Kruskal-Wallis and Mann-Whitney tests were applied to assess all semi-quantitative and quantitative variables. The correlation between each variable was also analyzed using Spearman rank at a p-value < 0.05. Results The results demonstrated that mice had different patterns of infiltrating cell types when compared to hamsters. This suggested that the cellular response to the infection in mice occurred earlier than that in hamsters. The response in mice reached its peak at D7 to D14 and then rapidly declined at D28. In contrast, although the inflammatory response in hamsters started slowly, the response reached the peak at D28 and maintained a high level until D56. Significant differences in the number of inflammatory cells between mice and hamsters were seen at D1 (p = 0.047), D7 (p = 0.049), D28 (p = 0.040), and D56 (p < 0.040). Conclusion The inflammatory responses to O. viverrini infection in the nonsusceptible animal model occurred and declined earlier while the response in the susceptible animal model occurred later in a gradual manner. Both rodents are suitable animal models for the studies of opisthorchiasis susceptibility.
Collapse
Affiliation(s)
- Theerayut Thongrin
- Faculty of Veterinary Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Watcharapol Suyapoh
- Faculty of Veterinary Science, Prince of Songkla University, Songkhla, Thailand
| | - WoroDanur Wendo
- Faculty of Veterinary Medicine, Khon Kaen University, Khon Kaen, Thailand
| | | | - Peerapol Sukon
- Faculty of Veterinary Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Kanin Salao
- Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Sutas Suttiprapa
- Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
- Tropical Disease Research Center, WHO Collaborating Centre for Research and Control of Opisthorchiasis, Khon Kaen University, Khon Kaen, Thailand
| | - Prasert Saichua
- Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
- Tropical Disease Research Center, WHO Collaborating Centre for Research and Control of Opisthorchiasis, Khon Kaen University, Khon Kaen, Thailand
| | - Sirikachorn Tangkawatana
- Faculty of Veterinary Medicine, Khon Kaen University, Khon Kaen, Thailand
- Tropical Disease Research Center, WHO Collaborating Centre for Research and Control of Opisthorchiasis, Khon Kaen University, Khon Kaen, Thailand
| |
Collapse
|
45
|
Summer M, Ali S, Fiaz U, Tahir HM, Ijaz M, Mumtaz S, Mushtaq R, Khan R, Shahzad H, Fiaz H. Therapeutic and immunomodulatory role of probiotics in breast cancer: A mechanistic review. Arch Microbiol 2023; 205:296. [PMID: 37486419 DOI: 10.1007/s00203-023-03632-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 07/03/2023] [Accepted: 07/09/2023] [Indexed: 07/25/2023]
Abstract
Breast cancer has become the most prevalent and noxious type of malignancy around the globe (Giaquinto et al., 2022). Multiple clinical strategies including chemotherapy, radiotherapy, and immunotherapy have been in practice to manage breast cancer. Besides the protective roles of conventional remedial approaches, and non-reversible and deteriorative impacts like healthy cell damage, organ failure, etc., the world scientific community is in a continuous struggle to find some alternative biocompatible and comparatively safe solutions. Among novel breast cancer management/treatment options, the role of probiotics has become immensely important. The current review encompasses the prevalence statistics of breast cancer across the globe concerning developed and undeveloped counties, intestinal microbiota linkage with breast cancer, and association of breast microbiome with breast carcinoma. Furthermore, this review also narrates the role of probiotics against breast cancer and their mode of action. In Vivo and In Vitro studies under breast cancer research regarding probiotics are mechanistically explained. The current review systematically explains the immunomodulatory role of probiotics to prevent breast cancer. Last, but not the least, current review concludes the use of probiotics in the treatment of breast cancer through various mechanisms and future recommendations for molecular basis studies.
Collapse
Affiliation(s)
- Muhammad Summer
- Department of Zoology, Government College University Lahore, Lahore, 54000, Pakistan
| | - Shaukat Ali
- Department of Zoology, Government College University Lahore, Lahore, 54000, Pakistan.
| | - Umaima Fiaz
- Department of Zoology, Government College University Lahore, Lahore, 54000, Pakistan
| | - Hafiz Muhammad Tahir
- Department of Zoology, Government College University Lahore, Lahore, 54000, Pakistan
| | - Muhammad Ijaz
- Department of Veterinary Medicine, University of Veterinary and Animal Sciences Lahore, Lahore, Pakistan
| | - Shumaila Mumtaz
- Department of Zoology, Government College University Lahore, Lahore, 54000, Pakistan
| | - Rabia Mushtaq
- Department of Zoology, Government College University Lahore, Lahore, 54000, Pakistan
| | - Rida Khan
- Department of Zoology, Government College University Lahore, Lahore, 54000, Pakistan
| | - Hafsa Shahzad
- Department of Zoology, Government College University Lahore, Lahore, 54000, Pakistan
| | - Hashim Fiaz
- Department of Medicine and Surgery, Ammer-ul-din Medical College Lahore, Lahore, Pakistan
| |
Collapse
|
46
|
Traserra S, Casabella-Ramón S, Vergara P, Jimenez M. E. coli infection disrupts the epithelial barrier and activates intrinsic neurosecretory reflexes in the pig colon. Front Physiol 2023; 14:1170822. [PMID: 37334046 PMCID: PMC10272729 DOI: 10.3389/fphys.2023.1170822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 05/24/2023] [Indexed: 06/20/2023] Open
Abstract
This study aims to assess the barrier integrity and possible activation of enteric neural pathways associated with secretion and motility in the pig colon induced by an enterotoxigenic Escherichia coli (ETEC) challenge. 50 Danbred male piglets were used for this study. 16 were challenged with an oral dose of the ETEC strain F4+ 1.5 × 109 colony-forming unit. Colonic samples were studied 4- and 9-days post-challenge using both a muscle bath and Ussing chamber. Colonic mast cells were stained with methylene blue. In control animals, electrical field stimulation induced neurosecretory responses that were abolished by tetrodotoxin (10-6M) and reduced by the combination of atropine (10-4M) and α-chymotrypsin (10U/mL). Exogenous addition of carbachol, vasoactive intestinal peptide, forskolin, 5-HT, nicotine, and histamine produced epithelial Cl- secretion. At day 4 post-challenge, ETEC increased the colonic permeability. The basal electrogenic ion transport remained increased until day 9 post-challenge and was decreased by tetrodotoxin (10-6M), atropine (10-4M), hexamethonium (10-5M), and ondansetron (10-5M). In the muscle, electrical field stimulation produced frequency-dependent contractile responses that were abolished with tetrodotoxin (10-6M) and atropine (10-6M). Electrical field stimulation and carbachol responses were not altered in ETEC animals in comparison with control animals at day 9 post-challenge. An increase in mast cells, stained with methylene blue, was observed in the mucosa and submucosa but not in the muscle layer of ETEC-infected animals on day 9 post-challenge. ETEC increased the response of intrinsic secretory reflexes and produced an impairment of the colonic barrier that was restored on day 9 post-challenge but did not modify neuromuscular function.
Collapse
Affiliation(s)
- Sara Traserra
- Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Sergi Casabella-Ramón
- Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Patri Vergara
- Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepaticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain
| | - Marcel Jimenez
- Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepaticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
47
|
Chen Y, Liu Z, Gong Y. Neuron-immunity communication: mechanism of neuroprotective effects in EGCG. Crit Rev Food Sci Nutr 2023; 64:9333-9352. [PMID: 37216484 DOI: 10.1080/10408398.2023.2212069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Epigallocatechin gallate (EGCG), a naturally occurring active ingredient unique to tea, has been shown to have neuroprotective potential. There is growing evidence of its potential advantages in the prevention and treatment of neuroinflammation, neurodegenerative diseases, and neurological damage. Neuroimmune communication is an important physiological mechanism in neurological diseases, including immune cell activation and response, cytokine delivery. EGCG shows great neuroprotective potential by modulating signals related to autoimmune response and improving communication between the nervous system and the immune system, effectively reducing the inflammatory state and neurological function. During neuroimmune communication, EGCG promotes the secretion of neurotrophic factors into the repair of damaged neurons, improves intestinal microenvironmental homeostasis, and ameliorates pathological phenotypes through molecular and cellular mechanisms related to the brain-gut axis. Here, we discuss the molecular and cellular mechanisms of inflammatory signaling exchange involving neuroimmune communication. We further emphasize that the neuroprotective role of EGCG is dependent on the modulatory role between immunity and neurology in neurologically related diseases.
Collapse
Affiliation(s)
- Ying Chen
- Key Laboratory of Tea Science of Ministry of Educatioxn, Changsha, China
- National Research Center of Engineering and Technology for Utilization of Botanical Functional Ingredients, Changsha, China
| | - Zhonghua Liu
- Key Laboratory of Tea Science of Ministry of Educatioxn, Changsha, China
- National Research Center of Engineering and Technology for Utilization of Botanical Functional Ingredients, Changsha, China
- Co-Innovation Center of Education Ministry for Utilization of Botanical Functional Ingredients, Changsha, China
- Key Laboratory for Evaluation and Utilization of Gene Resources of Horticultural Crops, Ministry of Agriculture and Rural Affairs of China, Hunan Agricultural University, Changsha, China
| | - Yushun Gong
- Key Laboratory of Tea Science of Ministry of Educatioxn, Changsha, China
- National Research Center of Engineering and Technology for Utilization of Botanical Functional Ingredients, Changsha, China
| |
Collapse
|
48
|
Van Kerschaver C, Turpin D, Michiels J, Pluske J. Reducing Weaning Stress in Piglets by Pre-Weaning Socialization and Gradual Separation from the Sow: A Review. Animals (Basel) 2023; 13:ani13101644. [PMID: 37238074 DOI: 10.3390/ani13101644] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 05/11/2023] [Accepted: 05/13/2023] [Indexed: 05/28/2023] Open
Abstract
The weaning of pigs in most commercial pork production systems is an abrupt event performed at a fairly young age, i.e., mostly between 2.5 and 5 weeks of age. This practice induces a stress response, and its impact on behavior, performance and the gastrointestinal tract has been well described. Historically, there has been a focus on pre- and post-weaning nutritional strategies and post-weaning housing conditions and medication to improve production and reduce mortality after weaning. However, alternative pre-weaning housing and management systems that promote the development of natural social behaviors of piglets before weaning have recently received more attention. Co-mingling of non-littermates before weaning is a strategy that aims to initiate social interactions prior to weaning. The separation of the litter from the sow in the period leading up to weaning, termed intermittent suckling, aims to enhance the gradual separation from the sow. In addition, these practices encourage the young pig to learn explorative nutrient sourcing. Altogether, they may reduce weaning-associated stress. In this review, these strategies are defined, and their effects on behavior, performance, mortality, gastrointestinal function and immunocompetence are described. Though these strategies may be adapted to a commercial setting, it also becomes clear that many factors can contribute to the success of these strategies.
Collapse
Affiliation(s)
- Céline Van Kerschaver
- Laboratory for Animal Nutrition and Animal Product Quality, Department of Animal Sciences and Aquatic Ecology, Ghent University, Coupure Links 653, 9000 Ghent, Belgium
| | - Diana Turpin
- Department of Primary Industries and Regional Development, 3 Baron-Hay Court, South Perth 6151, Australia
| | - Joris Michiels
- Laboratory for Animal Nutrition and Animal Product Quality, Department of Animal Sciences and Aquatic Ecology, Ghent University, Coupure Links 653, 9000 Ghent, Belgium
| | - John Pluske
- Australasian Pork Research Institute Limited, Willaston 5118, Australia
- Faculty of Science, The University of Melbourne, Parkville 3010, Australia
| |
Collapse
|
49
|
Sharkey KA, Mawe GM. The enteric nervous system. Physiol Rev 2023; 103:1487-1564. [PMID: 36521049 PMCID: PMC9970663 DOI: 10.1152/physrev.00018.2022] [Citation(s) in RCA: 136] [Impact Index Per Article: 68.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 12/12/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022] Open
Abstract
Of all the organ systems in the body, the gastrointestinal tract is the most complicated in terms of the numbers of structures involved, each with different functions, and the numbers and types of signaling molecules utilized. The digestion of food and absorption of nutrients, electrolytes, and water occurs in a hostile luminal environment that contains a large and diverse microbiota. At the core of regulatory control of the digestive and defensive functions of the gastrointestinal tract is the enteric nervous system (ENS), a complex system of neurons and glia in the gut wall. In this review, we discuss 1) the intrinsic neural control of gut functions involved in digestion and 2) how the ENS interacts with the immune system, gut microbiota, and epithelium to maintain mucosal defense and barrier function. We highlight developments that have revolutionized our understanding of the physiology and pathophysiology of enteric neural control. These include a new understanding of the molecular architecture of the ENS, the organization and function of enteric motor circuits, and the roles of enteric glia. We explore the transduction of luminal stimuli by enteroendocrine cells, the regulation of intestinal barrier function by enteric neurons and glia, local immune control by the ENS, and the role of the gut microbiota in regulating the structure and function of the ENS. Multifunctional enteric neurons work together with enteric glial cells, macrophages, interstitial cells, and enteroendocrine cells integrating an array of signals to initiate outputs that are precisely regulated in space and time to control digestion and intestinal homeostasis.
Collapse
Affiliation(s)
- Keith A Sharkey
- Hotchkiss Brain Institute and Snyder Institute for Chronic Diseases, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Gary M Mawe
- Department of Neurological Sciences, Larner College of Medicine, University of Vermont, Burlington, Vermont
| |
Collapse
|
50
|
Brucella abortus induces mast cell activation through TLR-2 and TLR-4. Microb Pathog 2023; 176:106005. [PMID: 36717005 DOI: 10.1016/j.micpath.2023.106005] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 01/23/2023] [Accepted: 01/25/2023] [Indexed: 01/29/2023]
Abstract
The Gram-negative bacteria Brucella abortus is a major cause of brucellosis in animals and humans. The host innate immune response to B. abortus is mainly associated with phagocytic cells such as dendritic cells, neutrophils, and macrophages. However, as mast cells naturally reside in the main bacterial entry sites they may be involved in bacterial recognition. At present, little is known about the role of mast cells during B. abortus infection. The role of the innate immune receptors TLR2 and TLR4 in activation of mast cells by B. abortus (strain RB51) infection was analyzed in this study. The results showed that B. abortus did not induce mast cell degranulation, but did induce the synthesis of the cytokines IL-1β, IL-6, TNF-α, CCL3, CCL4, and CCL5. Furthermore, B. abortus stimulated key cell signaling molecules involved in mast cell activation such as p38 and NF-κB. Blockade of the receptors TLR2 and TLR4 decreased TNF-α and IL-6 release by mast cells in response to B. abortus. Taken together, our results demonstrate that mast cells are activated by B. abortus and may play a role in inducing an inflammatory response during the initial phase of the infection.
Collapse
|