Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 1.772
Filtrer
1.
World J Gastroenterol ; 30(36): 4014-4020, 2024 Sep 28.
Article de Anglais | MEDLINE | ID: mdl-39351250

RÉSUMÉ

Gastrointestinal disorders encompass a spectrum of conditions affecting various organs within the digestive system, such as the esophagus, stomach, colon, rectum, pancreas, liver, small intestine, and bile ducts. The role of autophagy in the etiology and progression of gastrointestinal diseases has garnered significant attention. This paper seeks to evaluate the impact and mechanisms of autophagy in gastrointestinal disorders by synthesizing recent research findings. Specifically, we delve into inflammation-related gastrointestinal conditions, including ul-cerative colitis, Crohn's disease, and pancreatitis, as well as gastrointestinal cancers such as esophageal, gastric, and colorectal cancers. Additionally, we provide commentary on a recent publication by Chang et al in the World Journal of Gastroenterology. Our objective is to offer fresh perspectives on the mechanisms and therapeutic approaches for these gastrointestinal ailments. This review aims to offer new perspectives on the mechanisms and therapeutic strategies for gastrointestinal disorders by critically analyzing relevant publications. As discussed, the role of autophagy in gastrointestinal diseases is complex and, at times, contentious. To harness the full therapeutic potential of autophagy in treating these conditions, more in-depth research is imperative.


Sujet(s)
Autophagie , Maladies gastro-intestinales , Humains , Maladies gastro-intestinales/anatomopathologie , Maladies gastro-intestinales/physiopathologie , Maladies gastro-intestinales/thérapie , Tumeurs gastro-intestinales/anatomopathologie , Tumeurs gastro-intestinales/thérapie , Animaux , Tube digestif/immunologie , Tube digestif/anatomopathologie , Tube digestif/physiopathologie
2.
Gut Microbes ; 16(1): 2396494, 2024.
Article de Anglais | MEDLINE | ID: mdl-39340209

RÉSUMÉ

Bacterial extracellular vesicles (BEVs) are nano-sized lipid-shielded structures released by bacteria and that play an important role in intercellular communication. Their broad taxonomic origins and varying cargo compositions suggest their active participation in significant biological mechanisms. Specifically, they are involved in directly modulating microbial ecosystems, competing with other organisms, contributing to pathogenicity, and influencing the immunity of their hosts. This review examines the mechanisms that underlie the modulatory effects of BEVs on gut dynamics and immunity. Understanding how BEVs modulate microbiota composition and functional imbalances is crucial, as gut dysbiosis is implicated not only in the pathogenesis of various gastrointestinal, metabolic, and neurological diseases, but also in reducing resistance to colonization by enteric pathogens, which is particularly concerning given the current antimicrobial resistance crisis. This review summarizes recent advancements in the field of BEVs to encourage further research into these enigmatic entities. This will facilitate a better understanding of intra- and interkingdom communication phenomena and reveal promising therapeutic approaches.


Sujet(s)
Bactéries , Dysbiose , Vésicules extracellulaires , Microbiome gastro-intestinal , Vésicules extracellulaires/immunologie , Vésicules extracellulaires/métabolisme , Humains , Bactéries/immunologie , Bactéries/métabolisme , Bactéries/classification , Animaux , Dysbiose/microbiologie , Dysbiose/immunologie , Tube digestif/microbiologie , Tube digestif/immunologie
3.
Int J Mol Sci ; 25(17)2024 Aug 30.
Article de Anglais | MEDLINE | ID: mdl-39273369

RÉSUMÉ

The gut barrier is essential for protection against pathogens and maintaining homeostasis. Macrophages are key players in the immune system, are indispensable for intestinal health, and contribute to immune defense and repair mechanisms. Understanding the multifaceted roles of macrophages can provide critical insights into maintaining and restoring gastrointestinal (GI) health. This review explores the essential role of macrophages in maintaining the gut barrier function and their contribution to post-inflammatory and post-infectious responses in the gut. Macrophages significantly contribute to gut barrier integrity through epithelial repair, immune modulation, and interactions with gut microbiota. They demonstrate active plasticity by switching phenotypes to resolve inflammation, facilitate tissue repair, and regulate microbial populations following an infection or inflammation. In addition, tissue-resident (M2) and infiltration (M1) macrophages convert to each other in gut problems such as IBS and IBD via major signaling pathways mediated by NF-κB, JAK/STAT, PI3K/AKT, MAPK, Toll-like receptors, and specific microRNAs such as miR-155, miR-29, miR-146a, and miR-199, which may be good targets for new therapeutic approaches. Future research should focus on elucidating the detailed molecular mechanisms and developing personalized therapeutic approaches to fully harness the potential of macrophages to maintain and restore intestinal permeability and gut health.


Sujet(s)
Microbiome gastro-intestinal , Inflammation , Macrophages , Humains , Macrophages/immunologie , Macrophages/métabolisme , Animaux , Inflammation/métabolisme , Inflammation/immunologie , Muqueuse intestinale/métabolisme , Muqueuse intestinale/microbiologie , Muqueuse intestinale/immunologie , Transduction du signal , microARN/génétique , microARN/métabolisme , Tube digestif/microbiologie , Tube digestif/immunologie , Tube digestif/métabolisme , Perméabilité
6.
Arch Insect Biochem Physiol ; 116(4): e22130, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39118437

RÉSUMÉ

Toll receptors are important regulators of insects' innate immune system which, upon binding of pathogen molecules, activate a conserved signal transduction cascade known as the Toll pathway. RNA interference (RNAi) is a powerful tool to study the function of genes via reverse genetics. However, due to the reported refractory of RNAi efficiency in lepidopteran insects, successful reports of silencing of Toll receptors in the silkworm Bombyx mori have not been reported yet. In this study, a Toll receptor of the silkworm Bombyx Toll9-2 (BmToll9-2) was cloned and its expression and function were analyzed. The results showed that BmToll9-2 contains an ectodomain (ECD) with a signal peptide and nine leucine-rich repeats, a transmembrane helix, and a cytoplasmic region with a Toll/interleukin-1 domain. Phylogenetic analysis indicates that BmToll9-2 clusters with other insect Toll9 receptors and mammalian Toll-like receptor 4. Oral infection of exogenous pathogens showed that the Gram-negative bacterium Escherichia coli and its main cell wall component lipopolysaccharide (LPS), as well as the Gram-positive bacterium Staphylococcus aureus and its main cell wall component peptidoglycan, significantly induce BmToll9-2 expression in vivo. LPS also induced the expression of BmToll9-2 in BmN4 cells in vitro. These observations indicate its role as a sensor in the innate immunity to exogenous pathogens and as a pathogen-associated receptor that is responsive to LPS. RNAi of BmToll9-2 was effective in the midgut and epidermis. RNAi-mediated knock-down of BmToll9-2 reduced the weight and growth of the silkworm. Bacterial challenge following RNAi upregulated the expression of BmToll9-2 and rescued the weight differences of the silkworm, which may be related to its participation in the immune response and the regulation of the microbiota in the midgut lumen of the silkworm larvae.


Sujet(s)
Bombyx , Escherichia coli , Protéines d'insecte , Larve , Lipopolysaccharides , Phylogenèse , Animaux , Bombyx/immunologie , Bombyx/génétique , Bombyx/croissance et développement , Bombyx/microbiologie , Bombyx/métabolisme , Larve/immunologie , Larve/croissance et développement , Larve/microbiologie , Larve/génétique , Larve/métabolisme , Protéines d'insecte/génétique , Protéines d'insecte/métabolisme , Lipopolysaccharides/pharmacologie , Récepteurs de type Toll/métabolisme , Récepteurs de type Toll/génétique , Immunité innée , Staphylococcus aureus , Tube digestif/immunologie , Tube digestif/microbiologie , Tube digestif/métabolisme , Séquence d'acides aminés , Interférence par ARN
7.
Front Cell Infect Microbiol ; 14: 1450353, 2024.
Article de Anglais | MEDLINE | ID: mdl-39193502

RÉSUMÉ

Ticks are obligate hematophagous arthropods that transmit a wide range of pathogens to humans as well as wild and domestic animals. They also harbor a non-pathogenic microbiota, although our previous study has shown that the diverse bacterial microbiome in the midgut of Ixodes ricinus is quantitatively poor and lacks a core. In artificial infections by capillary feeding of ticks with two model bacteria (Gram-positive Micrococcus luteus and Gram-negative Pantoea sp.), rapid clearance of these microbes from the midgut was observed, indicating the presence of active immune mechanisms in this organ. In the current study, RNA-seq analysis was performed on the midgut of I. ricinus females inoculated with either M. luteus or Pantoea sp. or with sterile water as a control. While no immune-related transcripts were upregulated by microbial inoculation compared to that of the sterile control, capillary feeding itself triggered dramatic transcriptional changes in the tick midgut. Manual curation of the transcriptome from the midgut of unfed I. ricinus females, complemented by the proteomic analysis, revealed the presence of several constitutively expressed putative antimicrobial peptides (AMPs) that are independent of microbial stimulation and are referred to here as 'guard' AMPs. These included two types of midgut-specific defensins, two different domesticated amidase effector 2 (Dae2), microplusin/ricinusin-related molecules, two lysozymes, and two gamma interferon-inducible lysosomal thiol reductases (GILTs). The in vitro antimicrobial activity assays of two synthetic mature defensins, defensin 1 and defensin 8, confirmed their specificity against Gram-positive bacteria showing exceptional potency to inhibit the growth of M. luteus at nanomolar concentrations. The antimicrobial activity of midgut defensins is likely part of a multicomponent system responsible for the rapid clearance of bacteria in the tick midgut. Further studies are needed to evaluate the role of other identified 'guard' AMPs in controlling microorganisms entering the tick midgut.


Sujet(s)
Ixodes , Animaux , Ixodes/microbiologie , Ixodes/immunologie , Femelle , Micrococcus luteus/immunologie , Peptides antimicrobiens/métabolisme , Transcriptome , Tube digestif/microbiologie , Tube digestif/immunologie , Analyse de profil d'expression de gènes , Protéomique
8.
Immunol Rev ; 326(1): 66-82, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39046160

RÉSUMÉ

The increasing prevalence of food allergy and related pathologies in recent years has underscored the need to understand the factors affecting adverse reactions to food. Food allergy is caused when food-specific IgE triggers the release of histamine from mast cells. However, other food-specific antibody isotypes exist as well, including IgG and IgA. IgA is the main antibody isotype in the gut and mediates noninflammatory reactions to toxins, commensal bacteria, and food antigens. It has also been thought to induce tolerance to food, thus antagonizing the role of food-specific IgE. However, this has remained unclear as food-specific IgA generation is poorly understood. Particularly, the location of IgA induction, the role of T cell help, and the fates of food-specific B cells remain elusive. In this review, we outline what is known about food-specific IgA induction and highlight areas requiring further study. We also explore how knowledge of food-specific IgA induction can be informed by and subsequently contribute to our overall knowledge of gut immunity.


Sujet(s)
Hypersensibilité alimentaire , Immunoglobuline A , Humains , Animaux , Immunoglobuline A/immunologie , Immunoglobuline A/métabolisme , Hypersensibilité alimentaire/immunologie , Lymphocytes B/immunologie , Immunoglobuline E/immunologie , Immunoglobuline E/métabolisme , Immunité muqueuse , Lymphocytes T/immunologie , Tolérance immunitaire , Muqueuse intestinale/immunologie , Muqueuse intestinale/métabolisme , Tube digestif/immunologie , Microbiome gastro-intestinal/immunologie , Allergènes/immunologie
9.
J Dermatol ; 51(10): 1329-1334, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-38963343

RÉSUMÉ

This study aimed to identify severe gastrointestinal ailments in patients with systemic sclerosis (SSc), investigate the role of antibodies in gastrointestinal disorders, and explore the relationship between limited cutaneous SSc (lcSSc) and diffuse cutaneous SSc (dcSSc) in terms of gastrointestinal involvement and its association with skin stiffness. We used the University of California, Los Angeles, Scleroderma Clinical Trials Consortium Gastrointestinal Tract Instrument 2.0 (UCLA SCTC GIT 2.0) questionnaire to assess gastrointestinal disturbances in 100 patients with SSc. Gastrointestinal impairment was categorized into three levels: absence of or minor symptoms, moderate symptoms, and severe symptoms, as indicated by the total gastrointestinal tract (GIT) score. Comparing 27 patients with dcSSc and 73 patients with lcSSc, severe gastrointestinal disturbances were found in 7.4% of patients with dcSSc and 4.1% of patients with lcSSc. A total of 18.0% of anticentromere antibody (ACA)-positive patients exhibited moderate to severe symptoms, while 9.1% of antitopoisomerase 1 antibody-positive patients displayed similar symptoms. The average disease duration in patients with severe symptoms was 15.0 years, in those with moderate symptoms was 10.3 years, and in those who were symptom-free or mildly affected was 8.5 years. Among 16 patients with moderate to severe gastrointestinal disorders, a positive correlation was observed between the modified Rodnan skin thickness score (mRSS) and total GIT score. In addition, a positive correlation was identified between fecal incontinence and mRSS, with weaker correlations for reflux and bloating symptoms. Patients with gastrointestinal disorders showed a tendency to worsen over time, particularly in ACA-positive patients with dcSSc. Furthermore, a correlation was observed between mRSS and fecal incontinence, reflux, and abdominal bloating in patients with moderate to severe gastrointestinal disturbances.


Sujet(s)
Maladies gastro-intestinales , Indice de gravité de la maladie , Humains , Mâle , Femelle , Maladies gastro-intestinales/diagnostic , Maladies gastro-intestinales/étiologie , Adulte d'âge moyen , Études transversales , Adulte , Enquêtes et questionnaires/statistiques et données numériques , Sujet âgé , Sclérodermie systémique/complications , Sclérodermie systémique/diagnostic , Sclérodermie systémique/immunologie , Sclérodermie diffuse/complications , Sclérodermie diffuse/diagnostic , Sclérodermie diffuse/immunologie , Autoanticorps/sang , Autoanticorps/immunologie , Sclérodermie limitée/complications , Sclérodermie limitée/immunologie , Sclérodermie limitée/diagnostic , Tube digestif/immunologie , Tube digestif/imagerie diagnostique , ADN topoisomérases de type I/immunologie , Anticorps antinucléaires
10.
Int J Mol Sci ; 25(11)2024 May 29.
Article de Anglais | MEDLINE | ID: mdl-38892119

RÉSUMÉ

The immune system plays a key role in gastrointestinal (GI) pathologies, being responsible for protecting the body against infection, maintaining homeostasis, and regulating the inflammatory response in the GI tract [...].


Sujet(s)
Maladies gastro-intestinales , Système immunitaire , Humains , Maladies gastro-intestinales/immunologie , Système immunitaire/immunologie , Système immunitaire/métabolisme , Animaux , Homéostasie/immunologie , Tube digestif/immunologie , Tube digestif/métabolisme
11.
Curr Opin Crit Care ; 30(4): 290-297, 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-38872371

RÉSUMÉ

PURPOSE OF REVIEW: Gastrointestinal (GI) dysfunction limits enteral nutrition (EN) delivery in critical illness and contributes to systemic inflammation. The enteroendocrine (EE) axis plays an integral role in this interface between nutrition, inflammation, and GI function in critical illness. In this review, we present an overview of the EE system with a focus on its role in GI inflammation and function. RECENT FINDINGS: Enteroendocrine cells have been primarily described in their role in macronutrient digestion and absorption. Recent research has expanded on the diverse functions of EE cells including their ability to sense microbial peptides and metabolites and regulate immune function and inflammation. Therefore, EE cells may be both affected by and contribute to many pathophysiologic states and interventions of critical illness such as dysbiosis , inflammation, and alternative EN strategies. In this review, we present an overview of EE cells including their growing role in nonnutrient functions and integrate this understanding into relevant aspects of critical illness with a focus on EN. SUMMARY: The EE system is key in maintaining GI homeostasis in critical illness, and how it is impacted and contributes to outcomes in the setting of dysbiosis , inflammation and different feeding strategies in critical illness should be considered.


Sujet(s)
Maladie grave , Nutrition entérale , Cellules entéroendocrines , Inflammation , Humains , Inflammation/physiopathologie , Cellules entéroendocrines/physiologie , Dysbiose/physiopathologie , Tube digestif/physiopathologie , Tube digestif/immunologie , Tube digestif/physiologie , Microbiome gastro-intestinal/physiologie , Maladies gastro-intestinales/physiopathologie , État nutritionnel/physiologie
12.
Cell Immunol ; 401-402: 104844, 2024.
Article de Anglais | MEDLINE | ID: mdl-38901288

RÉSUMÉ

The gastrointestinal (GI) tract and the brain form bidirectional nervous, immune, and endocrine communications known as the gut-brain axis. Several factors can affect this axis; among them, various studies have focused on the microbiota and imply that alterations in microbiota combinations can influence both the brain and GI. Also, many studies have shown that the immune system has a vital role in varying gut microbiota combinations. In the current paper, we will review the multidirectional effects of gut microbiota, immune system, and nervous system on each other. Specifically, this review mainly focuses on the impact of Peyer's patches as a critical component of the gut immune system on the gut-brain axis through affecting the gut's microbial composition. In this way, some factors were discussed as proposed elements of missing gaps in this field.


Sujet(s)
Axe cerveau-intestin , Microbiome gastro-intestinal , Plaques de Peyer , Plaques de Peyer/immunologie , Humains , Microbiome gastro-intestinal/immunologie , Microbiome gastro-intestinal/physiologie , Animaux , Axe cerveau-intestin/physiologie , Axe cerveau-intestin/immunologie , Encéphale/immunologie , Encéphale/physiologie , Tube digestif/microbiologie , Tube digestif/immunologie
13.
Vet Q ; 44(1): 1-10, 2024 Dec.
Article de Anglais | MEDLINE | ID: mdl-38889341

RÉSUMÉ

The aim of the present study was to assess the effects of different levels of hempseed (HS) on growth performance, immunity and gut health in broiler chickens. A total of 192 Hubbard broiler chicks were divided into four groups and fed HS as follow: control (HS0), HS 10% (HS-10), HS 15% (HS-15) and HS 20% (HS-20). The study on HS supplementation in broilers revealed no significant impacts on feed intake during the starter (p = .2294) and finisher phases (p = .2294), or overall (p = .0944), though numerical increases were noted with higher HS levels. Body weight gain showed no significant influence in the starter and finisher phases, with overall weight gain also not significantly different (p = .0944), but numerically higher with increased HS. Feed conversion ratio was unaffected in the starter (p = .6986) and finisher phases (p = .6425), and overall (p = .2218). Dressing percentage (p = .1062) and mortality (p = .1631) were not significantly altered, but HS-20 had the highest dressing percentage and lowest mortality numerically. White blood cell counts increased significantly (p = .0377), especially in HS-15 and HS-20 groups. IgM and IgG production was higher in HS-20 on day 28 (p = .021). Gut pH (p > .05) and intestinal histomorphology (p > .05) were not significantly affected, although villus height increased numerically with higher HS levels. These results suggest potential benefits of HS, especially at higher inclusion levels. In conclusion, the obtained results indicated that HS incorporation into the diet of broilers did not affect the growth performance and gut health; however, the immune responses were significantly higher at 15 and 20% levels.


Sujet(s)
Aliment pour animaux , Cannabis , Poulets , Régime alimentaire , Compléments alimentaires , Animaux , Poulets/immunologie , Poulets/croissance et développement , Poulets/physiologie , Aliment pour animaux/analyse , Cannabis/composition chimique , Régime alimentaire/médecine vétérinaire , Phénomènes physiologiques nutritionnels chez l'animal/effets des médicaments et des substances chimiques , Mâle , Prise de poids/effets des médicaments et des substances chimiques , Tube digestif/effets des médicaments et des substances chimiques , Tube digestif/immunologie
14.
Curr Opin Immunol ; 87: 102425, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38763032

RÉSUMÉ

Interferons (IFNs) are an integral component of the host innate immune response during viral infection. Recent advances in the study of type I and III IFNs suggest that though both types counteract viral infection, type III IFNs act predominantly at epithelial barrier sites, while type I IFNs drive systemic responses. The dynamics and specific roles of type I versus III IFNs have been studied in the context of infection by a variety of enteric pathogens, including reovirus, rotavirus, norovirus, astrovirus, and intestinal severe acute respiratory syndrome coronavirus 2, revealing shared patterns of regulatory influence. An important role for the gut microbiota, including the virome, in regulating homeostasis and priming of intestinal IFN responses has also recently emerged.


Sujet(s)
Tube digestif , Interactions hôte-pathogène , Interféron lambda , Interféron de type I , Animaux , Humains , Microbiome gastro-intestinal/immunologie , Tube digestif/immunologie , Tube digestif/virologie , Tube digestif/microbiologie , Interactions hôte-pathogène/immunologie , Immunité innée , Interféron de type I/métabolisme , Interféron de type I/immunologie , Maladies virales/immunologie
15.
Trends Immunol ; 45(6): 470-481, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38782626

RÉSUMÉ

Skip lesions are an enigmatic spatial feature characterizing Crohn's disease (CD). They comprise inflamed and adjacent non-inflamed tissue sections with a clear demarcation. Currently, spatial features of the human gastrointestinal (GI) system lack clarity regarding the organization of microbes, mucus, tissue, and host cells during inflammation. New technologies with multiplexing abilities and innovative approaches provide ways of examining the spatial organization of inflamed and non-inflamed tissues in CD, which may open new avenues for diagnosis, prognosis, and treatment. In this review, we present evidence of the relevance of spatial context in patients with CD and the methods and ideas recently published in studies of spatiality during inflammation. With this review, we aim to provide inspiration for further research to address existing gaps.


Sujet(s)
Maladie de Crohn , Maladie de Crohn/immunologie , Maladie de Crohn/anatomopathologie , Humains , Animaux , Inflammation/immunologie , Muqueuse intestinale/immunologie , Muqueuse intestinale/anatomopathologie , Tube digestif/immunologie , Tube digestif/anatomopathologie
16.
Sci Adv ; 10(22): eadn7786, 2024 May 31.
Article de Anglais | MEDLINE | ID: mdl-38809992

RÉSUMÉ

Viruses, bacteria, and parasites frequently cause infections in the gastrointestinal tract, but traditional vaccination strategies typically elicit little or no mucosal antibody responses. Here, we report a strategy to effectively concentrate immunogens and adjuvants in gut-draining lymph nodes (LNs) to induce gut-associated mucosal immunity. We prepared nanoemulsions (NEs) based on biodegradable oils commonly used as vaccine adjuvants, which encapsulated a potent Toll-like receptor agonist and displayed antigen conjugated to their surface. Following intraperitoneal administration, these NEs accumulated in gut-draining mesenteric LNs, priming strong germinal center responses and promoting B cell class switching to immunoglobulin A (IgA). Optimized NEs elicited 10- to 1000-fold higher antigen-specific IgG and IgA titers in the serum and feces, respectively, compared to free antigen mixed with NE, and strong neutralizing antibody titers against severe acute respiratory syndrome coronavirus 2. Thus, robust gut humoral immunity can be elicited by exploiting the unique lymphatic collection pathways of the gut with a lymph-targeting vaccine formulation.


Sujet(s)
Immunité humorale , Animaux , Souris , Tube digestif/immunologie , Tissu lymphoïde/immunologie , Immunité muqueuse/effets des médicaments et des substances chimiques , SARS-CoV-2/immunologie , COVID-19/prévention et contrôle , COVID-19/immunologie , Anticorps antiviraux/immunologie , Noeuds lymphatiques/immunologie , Immunoglobuline A/immunologie , Vaccins contre la COVID-19/immunologie , Vaccins contre la COVID-19/administration et posologie , Anticorps neutralisants/immunologie , Femelle , Lymphocytes B/immunologie , Adjuvants vaccinaux , Souris de lignée C57BL , Humains
17.
Am J Physiol Gastrointest Liver Physiol ; 326(6): G712-G725, 2024 Jun 01.
Article de Anglais | MEDLINE | ID: mdl-38626403

RÉSUMÉ

Gut physiology is the epicenter of a web of internal communication systems (i.e., neural, immune, hormonal) mediated by cell-cell contacts, soluble factors, and external influences, such as the microbiome, diet, and the physical environment. Together these provide the signals that shape enteric homeostasis and, when they go awry, lead to disease. Faced with the seemingly paradoxical tasks of nutrient uptake (digestion) and retarding pathogen invasion (host defense), the gut integrates interactions between a variety of cells and signaling molecules to keep the host nourished and protected from pathogens. When the system fails, the outcome can be acute or chronic disease, often labeled as "idiopathic" in nature (e.g., irritable bowel syndrome, inflammatory bowel disease). Here we underscore the importance of a holistic approach to gut physiology, placing an emphasis on intercellular connectedness, using enteric neuroimmunophysiology as the paradigm. The goal of this opinion piece is to acknowledge the pace of change brought to our field via single-cell and -omic methodologies and other techniques such as cell lineage tracing, transgenic animal models, methods for culturing patient tissue, and advanced imaging. We identify gaps in the field and hope to inspire and challenge colleagues to take up the mantle and advance awareness of the subtleties, intricacies, and nuances of intestinal physiology in health and disease by defining communication pathways between gut resident cells, those recruited from the circulation, and "external" influences such as the central nervous system and the gut microbiota.


Sujet(s)
Microbiome gastro-intestinal , Tube digestif , Humains , Animaux , Tube digestif/immunologie , Tube digestif/physiologie , Tube digestif/microbiologie , Microbiome gastro-intestinal/physiologie , Neuro-immunomodulation/physiologie , Système nerveux entérique/physiologie , Système nerveux entérique/immunologie
18.
Autoimmun Rev ; 23(7-8): 103544, 2024.
Article de Anglais | MEDLINE | ID: mdl-38604462

RÉSUMÉ

Gut-tropic T cells primarily originate from gut-associated lymphoid tissue (GALT), and gut-tropic integrins mediate the trafficking of the T cells to the gastrointestinal tract, where their interplay with local hormones dictates the residence of the immune cells in both normal and compromised gastrointestinal tissues. Targeting gut-tropic integrins is an effective therapy for inflammatory bowel disease (IBD). Gut-tropic T cells are further capable of entering the peripheral circulatory system and relocating to multiple organs. There is mounting evidence indicating a correlation between gut-tropic T cells and extra-intestinal autoimmune disorders. This review aims to systematically discuss the origin, migration, and residence of gut-tropic T cells and their association with extra-intestinal autoimmune-related diseases. These discoveries are expected to offer new understandings into the development of a range of autoimmune disorders, as well as innovative approaches for preventing and treating the diseases.


Sujet(s)
Maladies auto-immunes , Lymphocytes T , Humains , Maladies auto-immunes/immunologie , Maladies auto-immunes/thérapie , Lymphocytes T/immunologie , Animaux , Tube digestif/immunologie , Maladies inflammatoires intestinales/immunologie , Maladies inflammatoires intestinales/thérapie
19.
Fish Shellfish Immunol ; 149: 109583, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38657879

RÉSUMÉ

Fish rely on mucosal surfaces as their first defence barrier against pathogens. Maintaining mucosal homeostasis is therefore crucial for their overall well-being, and it is likely that secreted immunoglobulins (sIg) play a pivotal role in sustaining this balance. In mammals, the poly-Ig receptor (pIgR) is an essential component responsible for transporting polymeric Igs across mucosal epithelia. In teleost fish, a counterpart of pIgR has been identified and characterized, exhibiting structural differences and broader mRNA expression patterns compared to mammals. Despite supporting evidence for the binding of Igs to recombinant pIgR proteins, the absence of a joining chain (J-chain) in teleosts challenges the conventional understanding of Ig transport mechanisms. The transport of IgM to the intestine via the hepatobiliary route is observed in vertebrates and has been proposed in a few teleosts. Investigations on the stomachless fish, ballan wrasse, revealed a significant role of the hepatobiliary route and interesting possibilities for alternative IgM transport routes that might include pancreatic tissue. These findings highlight the importance of gaining a thorough understanding of the mechanisms behind Ig transport to the gut in various teleosts. This review aims to gather existing information on pIgR-mediated transport across epithelial cells and immunoglobulin transport pathways to the gut lumen in teleost fish. It provides comparative insights into the hepatobiliary transport of Igs to the gut, emphasizing the current understanding in teleost fish while exploring potential alternative pathways for Ig transport to the gut lumen. Despite significant progress in understanding various aspects, there is still much to uncover, especially concerning the diversity of mechanisms across different teleost species.


Sujet(s)
Poissons , Immunoglobuline M , Animaux , Immunoglobuline M/immunologie , Poissons/immunologie , Poissons/génétique , Récepteur immunoglobuline polymérique/génétique , Récepteur immunoglobuline polymérique/immunologie , Récepteur immunoglobuline polymérique/composition chimique , Protéines de poisson/génétique , Protéines de poisson/immunologie , Protéines de poisson/composition chimique , Tube digestif/immunologie
20.
Immunity ; 57(5): 1019-1036.e9, 2024 May 14.
Article de Anglais | MEDLINE | ID: mdl-38677292

RÉSUMÉ

Group 3 innate lymphoid cells (ILC3) are the major subset of gut-resident ILC with essential roles in infections and tissue repair, but how they adapt to the gut environment to maintain tissue residency is unclear. We report that Tox2 is critical for gut ILC3 maintenance and function. Gut ILC3 highly expressed Tox2, and depletion of Tox2 markedly decreased ILC3 in gut but not at central sites, resulting in defective control of Citrobacter rodentium infection. Single-cell transcriptional profiling revealed decreased expression of Hexokinase-2 in Tox2-deficient gut ILC3. Consistent with the requirement for hexokinases in glycolysis, Tox2-/- ILC3 displayed decreased ability to utilize glycolysis for protein translation. Ectopic expression of Hexokinase-2 rescued Tox2-/- gut ILC3 defects. Hypoxia and interleukin (IL)-17A each induced Tox2 expression in ILC3, suggesting a mechanism by which ILC3 adjusts to fluctuating environments by programming glycolytic metabolism. Our results reveal the requirement for Tox2 to support the metabolic adaptation of ILC3 within the gastrointestinal tract.


Sujet(s)
Citrobacter rodentium , Infections à Enterobacteriaceae , Glycolyse , Protéines HMGB , Immunité innée , Lymphocytes , Souris knockout , Animaux , Souris , Adaptation physiologique/immunologie , Citrobacter rodentium/immunologie , Infections à Enterobacteriaceae/immunologie , Tube digestif/immunologie , Tube digestif/métabolisme , Hexokinase/métabolisme , Hexokinase/génétique , Interleukine-17/métabolisme , Lymphocytes/immunologie , Lymphocytes/métabolisme , Souris de lignée C57BL , Transactivateurs/métabolisme , Transactivateurs/génétique , Protéines HMGB/génétique , Protéines HMGB/immunologie , Protéines HMGB/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE