Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 12.587
Filtrer
1.
Vet Microbiol ; 295: 110154, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38959808

RÉSUMÉ

Porcine reproductive and respiratory syndrome (PRRS) is one of the costliest diseases to pork producers worldwide. We tested samples from the pregnant gilt model (PGM) to better understand the fetal response to in-utero PRRS virus (PRRSV) infection. Our goal was to identify critical tissues and genes associated with fetal resilience or susceptibility. Pregnant gilts (N=22) were infected with PRRSV on day 86 of gestation. At 21 days post maternal infection, the gilts and fetuses were euthanized, and fetal tissues collected. Fetuses were characterized for PRRS viral load in fetal serum and thymus, and preservation status (viable or meconium stained: VIA or MEC). Fetuses (N=10 per group) were compared: uninfected (UNIF; <1 log/µL PRRSV RNA), resilient (HV_VIA, >5 log virus/µL but viable), and susceptible (HV_MEC, >5 log virus/µL with MEC). Gene expression in fetal heart, kidney, and liver was investigated using NanoString transcriptomics. Gene categories investigated were hypothesized to be involved in fetal response to PRRSV infection: renin- angiotensin-aldosterone, inflammatory, transporter and metabolic systems. Following PRRSV infection, CCL5 increased expression in heart and kidney, and ACE2 decreased expression in kidney, each associated with fetal PRRS susceptibility. Liver revealed the most significant differential gene expression: CXCL10 decreased and IL10 increased indicative of immune suppression. Increased liver gene expression indicated potential associations with fetal PRRS susceptibility on several systems including blood pressure regulation (AGTR1), energy metabolism (SLC16A1 and SLC16A7), tissue specific responses (KL) and growth modulation (TGFB1). Overall, analyses of non-lymphoid tissues provided clues to mechanisms of fetal compromise following maternal PRRSV infection.


Sujet(s)
Résistance à la maladie , Foetus , Syndrome dysgénésique et respiratoire porcin , Transcriptome , Syndrome dysgénésique et respiratoire porcin/immunologie , Virus du syndrome respiratoire et reproducteur porcin/immunologie , Résistance à la maladie/génétique , Résistance à la maladie/immunologie , Grossesse , Animaux , Suidae , Femelle , Foetus/immunologie , Foetus/virologie , Régulation de l'expression des gènes/immunologie , Myocarde/immunologie , Foie/immunologie , Prédisposition aux maladies/immunologie , Complications infectieuses de la grossesse/immunologie , Complications infectieuses de la grossesse/médecine vétérinaire , Rein/immunologie
2.
Hepatol Commun ; 8(7)2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38967563

RÉSUMÉ

The liver is a vital organ that continuously adapts to a wide and dynamic diversity of self-antigens and xenobiotics. This involves the active contribution of immune cells, particularly by the liver-resident macrophages, the Kupffer cells (KCs), which exert a variety of central functions in liver homeostasis and disease. As such, KCs interact with their microenvironment to shape the hepatic cellular landscape, control gut-derived signal integration, and modulate metabolism. On injury, the rapid recruitment of bone marrow monocyte-derived macrophages alters this status quo and, when unrestrained, drastically compromises liver homeostasis, immune surveillance, and tissue organization. Several factors determine the functional roles of liver macrophages in these processes, such as their ontogeny, activation/polarization profile and, importantly, spatial distribution within the liver. Loss of tolerance and adaptability of the hepatic immune environment may result in persistent inflammation, hepatic fibrosis, cirrhosis, and a tumorigenic niche promoting liver cancer. In this review, we aim at providing the most recent breakthroughs in our understanding of liver macrophage biology, particularly their diversity and adaptability in the hepatic spatiotemporal context, as well as on potential therapeutic interventions that may hold the key to tackling remaining clinical challenges of varying etiologies in hepatology.


Sujet(s)
Cellules de Küpffer , Foie , Humains , Foie/immunologie , Foie/anatomopathologie , Cellules de Küpffer/immunologie , Cellules de Küpffer/physiologie , Animaux , Macrophages/immunologie , Macrophages/physiologie , Homéostasie/immunologie
3.
Int J Mol Sci ; 25(13)2024 Jul 07.
Article de Anglais | MEDLINE | ID: mdl-39000563

RÉSUMÉ

Circadian rhythms regulate physiological processes in approximately 24 h cycles, and their disruption is associated with various diseases. Inflammation may perturb circadian rhythms, though these interactions remain unclear. This study examined whether systemic inflammation induced by an intraperitoneal injection of lipopolysaccharide (LPS) could alter central and peripheral circadian rhythms and diurnal neuroimmune dynamics. Mice were randomly assigned to two groups: the saline control group and the LPS group. The diurnal expression of circadian clock genes and inflammatory cytokines were measured in the hypothalamus, hippocampus, and liver. Diurnal dynamic behaviors of microglia were also assessed. Our results revealed that the LPS perturbed circadian gene oscillations in the hypothalamus, hippocampus, and liver. Furthermore, systemic inflammation induced by the LPS could trigger neuroinflammation and perturb the diurnal dynamic behavior of microglia in the hippocampus. These findings shed light on the intricate link between inflammation and circadian disruption, underscoring their significance in relation to neurodegenerative diseases.


Sujet(s)
Rythme circadien , Inflammation , Lipopolysaccharides , Animaux , Souris , Mâle , Microglie/métabolisme , Microglie/immunologie , Hypothalamus/métabolisme , Hypothalamus/immunologie , Hippocampe/métabolisme , Cytokines/métabolisme , Foie/métabolisme , Foie/anatomopathologie , Foie/immunologie , Souris de lignée C57BL , Horloges circadiennes/génétique , Neuro-immunomodulation
4.
Adv Exp Med Biol ; 1445: 91-99, 2024.
Article de Anglais | MEDLINE | ID: mdl-38967752

RÉSUMÉ

Liver is the largest internal organ of the body with vital functions. In addition to its endocrine and exocrine activities, liver also plays a pivotal role in the immune system, including haematopoietic functions. Liver parenchymal cells, which are epithelial cells, have been found to possess innate immune functions by expressing pattern-recognition receptors (PRRs), producing complement components, and secreting cytokines. Intriguingly, in recent years, it has been discovered that liver epithelial cells also produce immunoglobulins (Igs), which have long been thought to be produced exclusively by B cells. Notably, even liver epithelial cells from B lymphocyte-deficient mice, including SCID mice and µMT mice, could also produce Igs. Compelling evidence has revealed both the physiological and pathological functions of liver-derived Igs. For instance, liver epithelial cells-derived IgM can serve as a source of natural and specific antibodies that contribute to innate immune responses, while liver-produced IgG can act as a growth factor to promote cell proliferation and survival in normal hepatocytes and hepatocarcinoma. Similar to that in B cells, the toll-like receptor 9 (TLR9)-MyD88 signaling pathway is also actively involved in promoting liver epithelial cells to secrete IgM. Liver-derived Igs could potentially serve as biomarkers, prognostic indicators, and therapeutic targets in the clinical setting, particularly for liver cancers and liver injury. Nevertheless, despite significant advances, much remains unknown about the mechanisms governing Ig transcription in liver cells, as well as the detailed functions of liver-derived Igs and their involvement in diseases and adaptive immunity. Further studies are still needed to reveal these underlying, undefined issues related to the role of liver-derived Igs in both immunity and diseases.


Sujet(s)
Immunité innée , Foie , Animaux , Foie/métabolisme , Foie/immunologie , Humains , Immunoglobulines/métabolisme , Immunoglobulines/immunologie , Immunoglobulines/génétique , Transduction du signal , Immunoglobuline M/immunologie , Immunoglobuline M/métabolisme , Tumeurs du foie/immunologie , Tumeurs du foie/anatomopathologie , Tumeurs du foie/métabolisme , Souris , Lymphocytes B/immunologie , Lymphocytes B/métabolisme , Hépatocytes/métabolisme , Hépatocytes/immunologie , Pertinence clinique
5.
J Immunol ; 213(3): 251-256, 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-39008791

RÉSUMÉ

Metabolic dysfunction-associated steatohepatitis (MASH) is characterized by infiltration of monocyte-derived macrophages (MdMs) into the liver; however, the function of these macrophages is largely unknown. We previously demonstrated that a population of MdMs, referred to as hepatic lipid-associated macrophages (LAMs), assemble into aggregates termed hepatic crown-like structures in areas of liver fibrosis. Intriguingly, decreasing MdM recruitment resulted in increased liver fibrosis, suggesting that LAMs contribute to antifibrotic pathways in MASH. In this study, we determined that hepatic crown-like structures are characterized by intimate interactions between activated hepatic stellate cells (HSCs) and macrophages in a collagen matrix in a mouse model of MASH. MASH macrophages displayed collagen-degrading capacities, and HSCs derived from MASH livers promoted expression of LAM marker genes and acquisition of a collagen-degrading phenotype in naive macrophages. These data suggest that crosstalk between HSCs and macrophages may contribute to collagen degradation MASH.


Sujet(s)
Cellules étoilées du foie , Cirrhose du foie , Macrophages , Phénotype , Cellules étoilées du foie/métabolisme , Cellules étoilées du foie/immunologie , Cellules étoilées du foie/anatomopathologie , Animaux , Souris , Macrophages/immunologie , Macrophages/métabolisme , Cirrhose du foie/anatomopathologie , Cirrhose du foie/métabolisme , Cirrhose du foie/immunologie , Souris de lignée C57BL , Collagène/métabolisme , Modèles animaux de maladie humaine , Humains , Foie/anatomopathologie , Foie/métabolisme , Foie/immunologie , Mâle , Stéatose hépatique/anatomopathologie , Stéatose hépatique/métabolisme , Stéatose hépatique/immunologie
6.
Sci Rep ; 14(1): 14919, 2024 06 28.
Article de Anglais | MEDLINE | ID: mdl-38942904

RÉSUMÉ

Helminth infections lead to an overdispersion of the parasites in humans as well as in animals. We asked whether early immune responses against migrating Ascaris larvae are responsible for the unequal distribution of worms in natural host populations and thus investigated a susceptible versus a resistant mouse strain. In mice, the roundworm larvae develop until the lung stage and thus early anti-Ascaris immune responses against the migrating larvae in the liver and lung can be deciphered. Our data show that susceptible C57BL/6 mice respond to Ascaris larval migration significantly stronger compared to resistant CBA mice and the anti-parasite reactivity is associated with pathology. Increased eosinophil recruitment was detected in the liver and lungs, but also in the spleen and peritoneal cavity of susceptible mice on day 8 post infection compared to resistant mice. In serum, eosinophil peroxidase levels were significantly higher only in the susceptible mice, indicating functional activity of the recruited eosinophils. This effect was associated with an increased IL-5/IL-13 production by innate lymphoid cells and CD4+ T cells and a pronounced type 2 macrophage polarization in the lungs of susceptible mice. Furthermore, a comparison of wildtype BALB/c and eosinophil-deficient dblGATA-1 BALB/c mice showed that eosinophils were not essential for the early control of migrating Ascaris larvae. In conclusion, in primary infection, a strong local and systemic type 2 immune response during hepato-tracheal helminth larval migration is associated with pathology rather than protection.


Sujet(s)
Ascaridiose , Larve , Poumon , Souris de lignée BALB C , Lymphocytes auxiliaires Th2 , Animaux , Ascaridiose/immunologie , Ascaridiose/parasitologie , Larve/immunologie , Souris , Lymphocytes auxiliaires Th2/immunologie , Poumon/parasitologie , Poumon/immunologie , Poumon/anatomopathologie , Ascaris/immunologie , Granulocytes éosinophiles/immunologie , Souris de lignée C57BL , Souris de lignée CBA , Foie/parasitologie , Foie/immunologie , Foie/anatomopathologie , Femelle
7.
Front Immunol ; 15: 1404891, 2024.
Article de Anglais | MEDLINE | ID: mdl-38919611

RÉSUMÉ

Background: Inflammatory cytokines play key pathogenic roles in liver fibrosis. IL-15 is a proinflammatory cytokine produced by myeloid cells. IL-15 promotes pathogenesis of several chronic inflammatory diseases. However, increased liver fibrosis has been reported in mice lacking IL-15 receptor alpha chain (IL-15Rα), suggesting an anti-fibrogenic role for IL-15. As myeloid cells are key players in liver fibrosis and IL-15 signaling can occur independently of IL-15Rα, we investigated the requirement of IL-15 and IL-15Rα in liver fibrosis. Methods: We induced liver fibrosis in Il15-/- , Il15ra-/- and wildtype C57BL/6 mice by the administration of carbon tetrachloride (CCl4). Liver fibrosis was evaluated by Sirius red and Mason's trichrome staining and α-smooth muscle acting immunostaining of myofibroblasts. Gene expression of collagens, matrix modifying enzymes, cytokines and chemokines was quantified by RT-qPCR. The phenotype and the numbers of intrahepatic lymphoid and myeloid cell subsets were evaluated by flow cytometry. Results: Both Il15-/- and Il15ra-/- mice developed markedly reduced liver fibrosis compared to wildtype control mice, as revealed by reduced collagen deposition and myofibroblast content. Il15ra-/- mice showed further reduction in collagen deposition compared to Il15-/- mice. However, Col1a1 and Col1a3 genes were similarly induced in the fibrotic livers of wildtype, Il15-/- and Il15ra-/- mice, although notable variations were observed in the expression of matrix remodeling enzymes and chemokines. As expected, Il15-/- and Il15ra-/- mice showed markedly reduced numbers of NK cells compared to wildtype mice. They also showed markedly less staining of CD45+ immune cells and CD68+ macrophages, and significantly reduced inflammatory cell infiltration into the liver, with fewer pro-inflammatory and anti-inflammatory monocyte subsets compared to wildtype mice. Conclusion: Our findings indicate that IL-15 exerts its profibrogenic role in the liver by promoting macrophage activation and that this requires trans-presentation of IL-15 by IL-15Rα.


Sujet(s)
Tétrachloro-méthane , Modèles animaux de maladie humaine , Sous-unité alpha du récepteur à l'interleukine-15 , Interleukine-15 , Cirrhose du foie , Souris de lignée C57BL , Souris knockout , Animaux , Interleukine-15/métabolisme , Interleukine-15/génétique , Souris , Sous-unité alpha du récepteur à l'interleukine-15/génétique , Sous-unité alpha du récepteur à l'interleukine-15/métabolisme , Cirrhose du foie/immunologie , Cirrhose du foie/anatomopathologie , Cirrhose du foie/métabolisme , Cirrhose du foie/induit chimiquement , Mâle , Foie/anatomopathologie , Foie/métabolisme , Foie/immunologie , Cytokines/métabolisme , Récepteurs à l'interleukine-15
8.
Cell Rep ; 43(6): 114324, 2024 Jun 25.
Article de Anglais | MEDLINE | ID: mdl-38850536

RÉSUMÉ

Trained immunity is classically characterized by long-term functional reprogramming of innate immune cells to combat infectious diseases. Infection-induced organ injury is a common clinical severity phenotype of sepsis. However, whether the induction of trained immunity plays a role in protecting septic organ injury remains largely unknown. Here, through establishing an in vivo ß-glucan training and lipopolysaccharide (LPS) challenge model in zebrafish larvae, we observe that induction of trained immunity could inhibit pyroptosis of hepatocytes to alleviate septic liver injury, with an elevated trimethyl-histone H3 lysine 4 (H3K4me3) modification that targets mitophagy-related genes. Moreover, we identify a C-type lectin domain receptor in zebrafish, named DrDectin-1, which is revealed as the orchestrator in gating H3K4me3 rewiring-mediated mitophagy activation and alleviating pyroptosis-engaged septic liver injury in vivo. Taken together, our results uncover tissue-resident trained immunity in maintaining liver homeostasis at the whole-animal level and offer an in vivo model to efficiently integrate trained immunity for immunotherapies.


Sujet(s)
Hépatocytes , Pyroptose , Sepsie , Protéines de poisson-zèbre , Danio zébré , Animaux , Hépatocytes/métabolisme , Hépatocytes/immunologie , Sepsie/immunologie , Protéines de poisson-zèbre/métabolisme , Protéines de poisson-zèbre/génétique , Lipopolysaccharides , Foie/anatomopathologie , Foie/métabolisme , Foie/immunologie , Mitophagie , Lectines de type C/métabolisme , Immunité innée , Histone/métabolisme , bêta-Glucanes/pharmacologie , Immunité entraînée
9.
Trends Immunol ; 45(7): 535-548, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38879436

RÉSUMÉ

Mammalian innate-like T cells (ILTCs), including mucosal-associated invariant T (MAIT), natural killer T (NKT), and γδ T cells, are abundant tissue-resident lymphocytes that have recently emerged as orchestrators of hepatic inflammation, tissue repair, and immune homeostasis. This review explores the involvement of different ILTC subsets in liver diseases. We explore the mechanisms underlying the pro- and anti-inflammatory effector functions of ILTCs in a context-dependent manner. We highlight latest findings regarding the dynamic interplay between ILTC functional subsets and other immune and parenchymal cells which may inform candidate immunomodulatory strategies to achieve improved clinical outcomes in liver diseases. We present new insights into how distinct gene expression programs in hepatic ILTCs are induced, maintained, and reprogrammed in a context- and disease stage-dependent manner.


Sujet(s)
Immunité innée , Maladies du foie , Humains , Animaux , Maladies du foie/immunologie , Cellules T invariantes associées aux muqueuses/immunologie , Cellules T tueuses naturelles/immunologie , Sous-populations de lymphocytes T/immunologie , Sous-populations de lymphocytes T/métabolisme , Foie/immunologie
10.
Genes (Basel) ; 15(6)2024 May 30.
Article de Anglais | MEDLINE | ID: mdl-38927648

RÉSUMÉ

Infections with gastrointestinal nematodes (GINs) reduce the economic efficiency of sheep operations and compromise animal welfare. Understanding the host's response to GIN infection can help producers identify animals that are naturally resistant to infection. The objective of this study was to characterize the hepatic transcriptome of sheep that had been naturally exposed to GIN parasites. The hepatic transcriptome was studied using RNA-Sequencing technology in animals characterized as high (n = 5) or medium (n = 6) based on their innate immune acute-phase (AP) response phenotype compared with uninfected controls (n = 4), and with biased antibody-mediated (AbMR, n = 5) or cell-mediated (CMR, n = 5) adaptive immune responsiveness compared to uninfected controls (n = 3). Following the assessment of sheep selected for innate responses, 0, 136, and 167 genes were differentially expressed (DE) between high- and medium-responding animals, high-responding and uninfected control animals, and medium-responding and uninfected control animals, respectively (false discovery rate (FDR) < 0.05, and fold change |FC| > 2). When adaptive immune responses were assessed, 0, 53, and 57 genes were DE between antibody- and cell-biased animals, antibody-biased and uninfected control animals, and cell-biased and uninfected control animals, respectively (FDR < 0.05, |FC| > 2). Functional analyses identified enriched gene ontology (GO) terms and metabolic pathways related to the innate immune response and energy metabolism. Six functional candidate genes were identified for further functional and validation studies to better understand the underlying biological mechanisms of host responses to GINs. These, in turn, can potentially help improve decision making and management practices to increase the overall host immune response to GIN infection.


Sujet(s)
Immunité innée , Foie , Nématodoses , Maladies des ovins , Transcriptome , Animaux , Ovis/parasitologie , Foie/parasitologie , Foie/métabolisme , Foie/immunologie , Nématodoses/médecine vétérinaire , Nématodoses/génétique , Nématodoses/immunologie , Nématodoses/parasitologie , Maladies des ovins/parasitologie , Maladies des ovins/génétique , Maladies des ovins/immunologie , Immunité innée/génétique , Nematoda/pathogénicité , Immunité acquise/génétique , Maladies gastro-intestinales/génétique , Maladies gastro-intestinales/parasitologie , Maladies gastro-intestinales/immunologie , Maladies gastro-intestinales/médecine vétérinaire
11.
J Nanobiotechnology ; 22(1): 315, 2024 Jun 05.
Article de Anglais | MEDLINE | ID: mdl-38840207

RÉSUMÉ

Chronic hepatitis B poses a significant global burden, modulating immune cells, leading to chronic inflammation and long-term damage. Due to its hepatotropism, the hepatitis B virus (HBV) cannot infect other cells. The mechanisms underlying the intercellular communication among different liver cells in HBV-infected individuals and the immune microenvironment imbalance remain elusive. Exosomes, as important intercellular communication and cargo transportation tools between HBV-infected hepatocytes and immune cells, have been shown to assist in HBV cargo transportation and regulate the immune microenvironment. However, the role of exosomes in hepatitis B has only gradually received attention in recent years. Minimal literature has systematically elaborated on the role of exosomes in reshaping the immune microenvironment of the liver. This review unfolds sequentially based on the biological processes of exosomes: exosomes' biogenesis, release, transport, uptake by recipient cells, and their impact on recipient cells. We delineate how HBV influences the biogenesis of exosomes, utilizing exosomal covert transmission, and reshapes the hepatic immune microenvironment. And based on the characteristics and functions of exosomes, potential applications of exosomes in hepatitis B are summarized and predicted.


Sujet(s)
Exosomes , Virus de l'hépatite B , Hépatite B chronique , Hépatocytes , Foie , Exosomes/immunologie , Exosomes/métabolisme , Humains , Virus de l'hépatite B/immunologie , Foie/immunologie , Foie/virologie , Animaux , Hépatite B chronique/immunologie , Hépatocytes/virologie , Hépatocytes/immunologie , Communication cellulaire , Microenvironnement cellulaire/immunologie , Hépatite B/immunologie , Hépatite B/virologie
12.
Cell Immunol ; 401-402: 104841, 2024.
Article de Anglais | MEDLINE | ID: mdl-38878619

RÉSUMÉ

Pneumonia persists as a public health crisis, representing the leading cause of death due to infection. Whether respiratory tract infections progress to pneumonia and its sequelae such as acute respiratory distress syndrome and sepsis depends on numerous underlying conditions related to both the causative agent and host. Regarding the former, pneumonia burden remains staggeringly high, despite the effectiveness of pathogen-targeting strategies such as vaccines and antibiotics. This demands a greater understanding of host features that collaborate to promote immune resistance and tissue resilience in the infected lung. Such features inside the pulmonary compartment have drawn much attention, where major advances have been made related to resident and recruited immune activity. By comparison, extra-pulmonary processes guiding pneumonia susceptibility are relatively elusive, constituting the focus of this review. Here we will highlight examples of when, how, and why tissues outside of the lungs dispatch signals that modulate local immunity in the airspaces. Topics include the liver, gut, bone marrow, brain and more, all of which contribute in direct and indirect ways to pneumonia outcome. When tuned appropriately, it has become clear that these responses can serve protective roles, and this will be considered distinctly from what would otherwise be aberrant responses characteristic of pneumonia-induced organ injury and sepsis. Further advances in this area may reveal novel targetable areas for clinical intervention that are not confined to the intra-pulmonary space.


Sujet(s)
Poumon , Humains , Animaux , Poumon/immunologie , Pneumopathie infectieuse/immunologie , Foie/immunologie , Infections de l'appareil respiratoire/immunologie , Encéphale/immunologie , Sepsie/immunologie
13.
Arch Virol ; 169(7): 148, 2024 Jun 18.
Article de Anglais | MEDLINE | ID: mdl-38888759

RÉSUMÉ

The inflammasome is a multimeric protein complex that plays a vital role in the defence against pathogens and is therefore considered an essential component of the innate immune system. In this study, the expression patterns of inflammasome genes (NLRC3, ASC, and CAS-1), antiviral genes (IFNγ and MX), and immune genes (IL-1ß and IL-18) were analysed in Oreochromis niloticus liver (ONIL) cells following stimulation with the bacterial ligands peptidoglycan (PGN) and lipopolysaccharide (LPS) and infection with TiLV. The cells were stimulated with PGN and LPS at concentrations of 10, 25, and 50 µg/ml. For viral infection, 106 TCID50 of TiLV per ml was used. After LPS stimulation, all seven genes were found to be expressed at specific time points at each of the three doses tested. However, at even higher doses of LPS, NLRC3 levels decreased. Following TiLV infection, all of the genes showed significant upregulation, especially at early time points. However, the gene expression pattern was found to be unique in PGN-treated cells. For instance, NLRC3 and ASC did not show any response to PGN stimulation, and the expression of IFNγ was downregulated at 25 and 50 µg of PGN per ml. CAS-1 and IL-18 expression was downregulated at 25 µg of PGN per ml. At a higher dose (50 µg/ml), IL-1ß showed downregulation. Overall, our results indicate that these genes are involved in the immune response to viral and bacterial infection and that the degree of response is ligand- and dose-dependent.


Sujet(s)
Cichlides , Maladies des poissons , Inflammasomes , Animaux , Cichlides/immunologie , Cichlides/génétique , Inflammasomes/génétique , Inflammasomes/immunologie , Inflammasomes/métabolisme , Maladies des poissons/immunologie , Maladies des poissons/virologie , Maladies des poissons/microbiologie , Maladies des poissons/génétique , Lignée cellulaire , Peptidoglycane/pharmacologie , Foie/virologie , Foie/immunologie , Lipopolysaccharides/pharmacologie , Immunité innée , Protéines de poisson/génétique , Interleukine-18/génétique , Interleukine-18/métabolisme , Ligands , Infections à virus à ADN/immunologie , Infections à virus à ADN/médecine vétérinaire , Infections à virus à ADN/virologie , Infections à virus à ADN/génétique , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Régulation de l'expression des gènes/immunologie , Interleukine-1 bêta/génétique , Interleukine-1 bêta/métabolisme , Interleukine-1 bêta/immunologie
14.
Int Immunopharmacol ; 137: 112442, 2024 Aug 20.
Article de Anglais | MEDLINE | ID: mdl-38889508

RÉSUMÉ

Liver disease ranks as the eleventh leading cause of mortality, leading to approximately 2 million deaths annually worldwide. Neutrophils are a type of immune cell that are abundant in peripheral blood and play a vital role in innate immunity by quickly reaching the site of liver injury. They exert their influence on liver diseases through autocrine, paracrine, and immunomodulatory mechanisms. Extracellular vesicles, phospholipid bilayer vesicles, transport a variety of substances, such as proteins, nucleic acids, lipids, and pathogenic factors, for intercellular communication. They regulate cell communication and perform their functions by delivering biological information. Current research has revealed the involvement of the interaction between neutrophils and extracellular vesicles in the pathogenesis of liver disease. Moreover, more research has focused on targeting neutrophils as a therapeutic strategy to attenuate disease progression. Therefore, this article summarizes the roles of neutrophils, extracellular vesicles, and their interactions in noncancerous liver diseases.


Sujet(s)
Communication cellulaire , Vésicules extracellulaires , Maladies du foie , Granulocytes neutrophiles , Humains , Vésicules extracellulaires/métabolisme , Vésicules extracellulaires/immunologie , Granulocytes neutrophiles/immunologie , Granulocytes neutrophiles/métabolisme , Maladies du foie/immunologie , Maladies du foie/métabolisme , Maladies du foie/anatomopathologie , Animaux , Communication cellulaire/immunologie , Immunité innée , Foie/métabolisme , Foie/anatomopathologie , Foie/immunologie
15.
Int Immunopharmacol ; 137: 112289, 2024 Aug 20.
Article de Anglais | MEDLINE | ID: mdl-38889505

RÉSUMÉ

Fms-like tyrosine kinase 3 (FLT3) is a receptor tyrosine kinase (RTK) primarily expressed in hematopoietic stem cells and dendritic cells (DCs). While FLT3 plays a critical role in the proliferation, development and maintenance of DCs, thus influencing immune responses under both normal and pathological conditions, there also exists some evidence that FLT3+DC may be involved with immune responses in liver transplantation (LT). In this study, results from single-cell sequencing data analysis revealed a clear relationship between FLT3+DCs and Regulatory T cells (Tregs) in liver tissue of LT recipients. In peripheral blood samples of LT patients, levels of FLT3+DCs were decreased post-LT-surgery, while Tregs were increased. In a LT mouse model, levels of FLT3+DCs in the liver and bone marrow exhibited an initial time-dependent decrease followed by an increase after LT surgery. Results as obtained with co-culture experiments using mature BMDCs and CD4+ T cells revealed fluctuations in Tregs in response to FLT3 inhibitors and the FLT3 ligand. These findings suggest that FLT3+DCs could emerge as a novel target for mitigating immune rejection in LT.


Sujet(s)
Cellules dendritiques , Rejet du greffon , Transplantation hépatique , Souris de lignée C57BL , Lymphocytes T régulateurs , Tyrosine kinase-3 de type fms , Lymphocytes T régulateurs/immunologie , Animaux , Cellules dendritiques/immunologie , Tyrosine kinase-3 de type fms/métabolisme , Humains , Rejet du greffon/immunologie , Rejet du greffon/prévention et contrôle , Mâle , Souris , Foie/immunologie , Femelle , Techniques de coculture , Adulte d'âge moyen , Cellules cultivées , Souris de lignée BALB C , Protéines membranaires
16.
Front Immunol ; 15: 1381735, 2024.
Article de Anglais | MEDLINE | ID: mdl-38840923

RÉSUMÉ

Background: Acute liver injury (ALI), which is a type of inflammation-mediated hepatocellular injury, is a clinical syndrome that results from hepatocellular apoptosis and hemorrhagic necrosis. Apoptosis stimulating protein of p53-2 (ASPP2) is a proapoptotic member of the p53 binding protein family. However, the role of ASPP2 in the pathogenesis of ALI and its regulatory mechanisms remain unclear. Methods: The expression of ASPP2 were compared between liver biopsies derived from patients with CHB, patients with ALI, and normal controls. Acute liver injury was modelled in mice by administration of D-GalN/LPS. Liver injury was demonstrated by serum transaminases and histological assessment of liver sections. ASPP2-knockdown mice (ASPP2+/-) were used to determine its role in acute liver injury. Mouse bone marrow macrophages (BMMs) were isolated from wildtype and ASPP2+/- mice and stimulated with LPS, and the supernatant was collected to incubate with the primary hepatocytes. Quantitative real-time PCR and western blot were used to analyze the expression level of target. Results: The expression of ASPP2 was significantly upregulated in the liver tissue of ALI patients and acute liver injury mice. ASPP2+/- mice significantly relieved liver injury through reducing liver inflammation and decreasing hepatocyte apoptosis. Moreover, the conditioned medium (CM) of ASPP2+/- bone marrow-derived macrophages (BMMs) protected hepatocytes against apoptosis. Mechanistically, we revealed that ASPP2 deficiency in BMMs specifically upregulated IL-6 through autophagy activation, which decreased the level of TNF-α to reduce hepatocytes apoptosis. Furthermore, up-regulation of ASPP2 sensitizes hepatocytes to TNF-α-induced apoptosis. Conclusion: Our novel findings show the critical role of ASPP2 in inflammatory immunoregulatory mechanism of ALI and provide a rationale to target ASPP2 as a refined therapeutic strategy to ameliorate acute liver injury.


Sujet(s)
Protéines régulatrices de l'apoptose , Apoptose , Animaux , Humains , Souris , Mâle , Protéines régulatrices de l'apoptose/métabolisme , Protéines régulatrices de l'apoptose/génétique , Souris knockout , Foie/anatomopathologie , Foie/métabolisme , Foie/immunologie , Hépatocytes/métabolisme , Hépatocytes/anatomopathologie , Souris de lignée C57BL , Modèles animaux de maladie humaine , Inflammation/immunologie , Inflammation/métabolisme , Femelle , Lipopolysaccharides , Adulte d'âge moyen , Macrophages/immunologie , Macrophages/métabolisme , Adulte , Protéines suppresseurs de tumeurs
17.
Hepatol Commun ; 8(7)2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38896080

RÉSUMÉ

BACKGROUND: Neutrophils are key mediators of inflammation during acute liver injury (ALI). Emerging evidence suggests that they also contribute to injury resolution and tissue repair. However, the different neutrophil subsets involved in these processes and their kinetics are undefined. Herein, we characterized neutrophil kinetics and heterogeneity during ALI. METHODS: We used the carbon tetrachloride model of ALI and employed flow cytometry, tissue imaging, and quantitative RT-PCR to characterize intrahepatic neutrophils during the necroinflammatory early and late repair phases of the wound healing response to ALI. We FACS sorted intrahepatic neutrophils at key time points and examined their transcriptional profiles using RNA-sequencing. Finally, we evaluated neutrophil protein translation, mitochondrial function and metabolism, reactive oxygen species content, and neutrophil extracellular traps generation. RESULTS: We detected 2 temporarily distinct waves of neutrophils during (1) necroinflammation (at 24 hours after injury) and (2) late repair (at 72 hours). Early neutrophils were proinflammatory, characterized by: (1) upregulation of inflammatory cytokines, (2) activation of the noncanonical NF-κB pathway, (3) reduction of protein translation, (4) decreased oxidative phosphorylation, and (5) higher propensity to generate reactive oxygen species and neutrophil extracellular traps. In contrast, late neutrophils were prorepair and enriched in genes and pathways associated with tissue repair and angiogenesis. Finally, early proinflammatory neutrophils were characterized by the expression of a short isoform of C-X-C chemokine receptor 5, while the late prorepair neutrophils were characterized by the expression of C-X-C chemokine receptor 4. CONCLUSIONS: This study underscores the phenotypic and functional heterogeneity of neutrophils and their dual role in inflammation and tissue repair during ALI.


Sujet(s)
Granulocytes neutrophiles , Animaux , Granulocytes neutrophiles/immunologie , Granulocytes neutrophiles/métabolisme , Souris , Modèles animaux de maladie humaine , Souris de lignée C57BL , Mâle , Espèces réactives de l'oxygène/métabolisme , Foie/anatomopathologie , Foie/immunologie , Lésions hépatiques dues aux substances/immunologie , Lésions hépatiques dues aux substances/génétique , Cytokines/métabolisme , Pièges extracellulaires/métabolisme
18.
Int J Mol Sci ; 25(11)2024 Jun 05.
Article de Anglais | MEDLINE | ID: mdl-38892423

RÉSUMÉ

The autonomic nervous system plays an integral role in motion and sensation as well as the physiologic function of visceral organs. The nervous system additionally plays a key role in primary liver diseases. Until recently, however, the impact of nerves on cancer development, progression, and metastasis has been unappreciated. This review highlights recent advances in understanding neuroanatomical networks within solid organs and their mechanistic influence on organ function, specifically in the liver and liver cancer. We discuss the interaction between the autonomic nervous system, including sympathetic and parasympathetic nerves, and the liver. We also examine how sympathetic innervation affects metabolic functions and diseases like nonalcoholic fatty liver disease (NAFLD). We also delve into the neurobiology of the liver, the interplay between cancer and nerves, and the neural regulation of the immune response. We emphasize the influence of the neuroimmune axis in cancer progression and the potential of targeted interventions like neurolysis to improve cancer treatment outcomes, especially for hepatocellular carcinoma (HCC).


Sujet(s)
Carcinome hépatocellulaire , Tumeurs du foie , Neuro-immunomodulation , Humains , Tumeurs du foie/immunologie , Tumeurs du foie/thérapie , Tumeurs du foie/anatomopathologie , Tumeurs du foie/métabolisme , Animaux , Carcinome hépatocellulaire/immunologie , Carcinome hépatocellulaire/thérapie , Carcinome hépatocellulaire/anatomopathologie , Carcinome hépatocellulaire/métabolisme , Foie/anatomopathologie , Foie/immunologie , Foie/métabolisme , Stéatose hépatique non alcoolique/immunologie , Stéatose hépatique non alcoolique/thérapie , Stéatose hépatique non alcoolique/anatomopathologie , Stéatose hépatique non alcoolique/métabolisme , Système nerveux autonome/physiopathologie
19.
Parasites Hosts Dis ; 62(2): 243-250, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38835265

RÉSUMÉ

We investigated organ specific Toxocara canis larval migration in mice infected with T. canis larvae. We observed the worm burden and systemic immune responses. Three groups of BALB/c mice (n=5 each) were orally administered 1,000 T. canis 2nd stage larvae to induce larva migrans. Mice were sacrificed at 1, 3, and 5 weeks post-infection. Liver, lung, brain, and eye tissues were collected. Tissue from 2 mice per group was digested for larval count, while the remaining 3 mice underwent histological analysis. Blood hematology and serology were evaluated and compared to that in a control uninfected group (n=5) to assess the immune response. Cytokine levels in bronchoalveolar lavage (BAL) fluid were also analyzed. We found that, 1 week post-infection, the mean parasite load in the liver (72±7.1), brain (31±4.2), lungs (20±5.7), and eyes (2±0) peaked and stayed constant until the 3 weeks. By 5-week post-infection, the worm burden in the liver and lungs significantly decreased to 10±4.2 and 9±5.7, respectively, while they remained relatively stable in the brain and eyes (18±4.2 and 1±0, respectively). Interestingly, ocular larvae resided in all retinal layers, without notable inflammation in outer retina. Mice infected with T. canis exhibited elevated levels of neutrophils, monocytes, eosinophils, and immunoglobulin E. At 5 weeks post-infection, interleukin (IL)-5 and IL-13 levels were elevated in BAL fluid. Whereas IL-4, IL-10, IL-17, and interferon-γ levels in BAL fluid were similar to that in controls. Our findings demonstrate that a small portion of T. canis larvae migrate to the eyes and brain within the first week of infection. Minimal tissue inflammation was observed, probably due to increase of anti-inflammatory cytokines. This study contributes to our understanding of the histological and immunological responses to T. canis infection in mice, which may have implications to further understand human toxocariasis.


Sujet(s)
Encéphale , Cytokines , Larve , Foie , Poumon , Souris de lignée BALB C , Toxocara canis , Toxocarose , Animaux , Toxocara canis/immunologie , Toxocarose/immunologie , Toxocarose/anatomopathologie , Toxocarose/parasitologie , Larve/immunologie , Souris , Cytokines/métabolisme , Poumon/parasitologie , Poumon/immunologie , Poumon/anatomopathologie , Foie/parasitologie , Foie/anatomopathologie , Foie/immunologie , Encéphale/parasitologie , Encéphale/immunologie , Encéphale/anatomopathologie , Liquide de lavage bronchoalvéolaire/immunologie , Liquide de lavage bronchoalvéolaire/parasitologie , Femelle , Charge parasitaire , Oeil/parasitologie , Oeil/immunologie , Oeil/anatomopathologie , Modèles animaux de maladie humaine
20.
Nat Commun ; 15(1): 4711, 2024 Jun 03.
Article de Anglais | MEDLINE | ID: mdl-38830841

RÉSUMÉ

The fetal development of organs and functions is vulnerable to perturbation by maternal inflammation which may increase susceptibility to disorders after birth. Because it is not well understood how the placenta and fetus respond to acute lung- inflammation, we characterize the response to maternal pulmonary lipopolysaccharide exposure across 24 h in maternal and fetal organs using multi-omics, imaging and integrative analyses. Unlike maternal organs, which mount strong inflammatory immune responses, the placenta upregulates immuno-modulatory genes, in particular the IL-6 signaling suppressor Socs3. Similarly, we observe no immune response in the fetal liver, which instead displays metabolic changes, including increases in lipids containing docosahexaenoic acid, crucial for fetal brain development. The maternal liver and plasma display similar metabolic alterations, potentially increasing bioavailability of docosahexaenoic acid for the mother and fetus. Thus, our integrated temporal analysis shows that systemic inflammation in the mother leads to a metabolic perturbation in the fetus.


Sujet(s)
Foetus , Lipopolysaccharides , Foie , Poumon , Placenta , Femelle , Grossesse , Placenta/métabolisme , Placenta/immunologie , Animaux , Foetus/immunologie , Foetus/métabolisme , Poumon/immunologie , Poumon/métabolisme , Foie/métabolisme , Foie/immunologie , Acide docosahexaénoïque/métabolisme , Protéine-3 suppressive de la signalisation des cytokine/métabolisme , Protéine-3 suppressive de la signalisation des cytokine/génétique , Souris , Inflammation/immunologie , Inflammation/métabolisme , Souris de lignée C57BL , Adaptation physiologique/immunologie , Développement foetal/immunologie , Échange foetomaternel/immunologie , Interleukine-6/métabolisme , Interleukine-6/immunologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...