Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 354
Filter
1.
Immunity ; 54(12): 2795-2811.e9, 2021 12 14.
Article in English | MEDLINE | ID: mdl-34788601

ABSTRACT

Lymphangitis and the formation of tertiary lymphoid organs (TLOs) in the mesentery are features of Crohn's disease. Here, we examined the genesis of these TLOs and their impact on disease progression. Whole-mount and intravital imaging of the ileum and ileum-draining collecting lymphatic vessels (CLVs) draining to mesenteric lymph nodes from TNFΔARE mice, a model of ileitis, revealed TLO formation at valves of CLVs. TLOs obstructed cellular and molecular outflow from the gut and were sites of lymph leakage and backflow. Tumor necrosis factor (TNF) neutralization begun at early stages of TLO formation restored lymph transport. However, robustly developed, chronic TLOs resisted regression and restoration of flow after TNF neutralization. TNF stimulation of cultured lymphatic endothelial cells reprogrammed responses to oscillatory shear stress, preventing the induction of valve-associated genes. Disrupted transport of immune cells, driven by loss of valve integrity and TLO formation, may contribute to the pathology of Crohn's disease.


Subject(s)
Crohn Disease/immunology , Endothelial Cells/immunology , Ileum/immunology , Lymph/metabolism , Lymphatic Vessels/immunology , Mesentery/immunology , Tertiary Lymphoid Structures/immunology , Tumor Necrosis Factor-alpha/metabolism , Animals , Cell Movement , Cells, Cultured , Disease Models, Animal , Humans , Ileitis , Lymphangitis , Mice , Mice, Knockout , Stress, Mechanical
2.
Cell ; 163(2): 354-66, 2015 Oct 08.
Article in English | MEDLINE | ID: mdl-26451485

ABSTRACT

Infections have been proposed as initiating factors for inflammatory disorders; however, identifying associations between defined infectious agents and the initiation of chronic disease has remained elusive. Here, we report that a single acute infection can have dramatic and long-term consequences for tissue-specific immunity. Following clearance of Yersinia pseudotuberculosis, sustained inflammation and associated lymphatic leakage in the mesenteric adipose tissue deviates migratory dendritic cells to the adipose compartment, thereby preventing their accumulation in the mesenteric lymph node. As a consequence, canonical mucosal immune functions, including tolerance and protective immunity, are persistently compromised. Post-resolution of infection, signals derived from the microbiota maintain inflammatory mesentery remodeling and consequently, transient ablation of the microbiota restores mucosal immunity. Our results indicate that persistent disruption of communication between tissues and the immune system following clearance of an acute infection represents an inflection point beyond which tissue homeostasis and immunity is compromised for the long-term. VIDEO ABSTRACT.


Subject(s)
Gastrointestinal Microbiome , Immune System Diseases/microbiology , Immune System Diseases/pathology , Lymphatic Diseases/pathology , Yersinia pseudotuberculosis Infections/immunology , Yersinia pseudotuberculosis/physiology , Cell Movement , Chronic Disease , Dendritic Cells/pathology , Female , Humans , Lymphatic Diseases/microbiology , Lymphoid Tissue/immunology , Lymphoid Tissue/pathology , Male , Mesentery/immunology , Mesentery/pathology , Specific Pathogen-Free Organisms , Yersinia pseudotuberculosis Infections/pathology
3.
J Immunol ; 204(5): 1075-1083, 2020 03 01.
Article in English | MEDLINE | ID: mdl-32071090

ABSTRACT

The mammalian intestine is a complex environment that is constantly exposed to Ags derived from food, microbiota, and metabolites. Intestinal dendritic cells (DC) have the responsibility of establishing oral tolerance against these Ags while initiating immune responses against mucosal pathogens. We now know that DC are a heterogeneous population of innate immune cells composed of classical and monocyte-derived DC, Langerhans cells, and plasmacytoid DC. In the intestine, DC are found in organized lymphoid tissues, such as the mesenteric lymph nodes and Peyer's patches, as well as in the lamina propria. In this Brief Review, we review recent work that describes a division of labor between and collaboration among gut DC subsets in the context of intestinal homeostasis and inflammation. Understanding relationships between DC subtypes and their biological functions will rationalize oral vaccine design and will provide insights into treatments that quiet pathological intestinal inflammation.


Subject(s)
Immunity, Mucosal , Intestinal Mucosa/immunology , Langerhans Cells/immunology , Peyer's Patches/immunology , Animals , Humans , Inflammation/immunology , Inflammation/pathology , Intestinal Mucosa/pathology , Langerhans Cells/pathology , Lymph Nodes/immunology , Lymph Nodes/pathology , Mesentery/immunology , Mesentery/pathology , Peyer's Patches/pathology
4.
J Immunol ; 204(7): 1859-1868, 2020 04 01.
Article in English | MEDLINE | ID: mdl-32122996

ABSTRACT

Dendritic cells (DCs) participate in the pathogenesis of several diseases. We investigated DCs and the connection between mucosa and joints in a murine model of Yersinia enterocolitica O:3-induced reactive arthritis (ReA) in TNFRp55-/- mice. DCs of mesenteric lymph nodes (MLN) and joint regional lymph nodes (RLN) were analyzed in TNFRp55-/- and wild-type mice. On day 14 after Y. enterocolitica infection (arthritis onset), we found that under TNFRp55 deficiency, migratory (MHChighCD11c+) DCs increased significantly in RLN. Within these RLN, resident (MHCintCD11c+) DCs increased on days 14 and 21. Similar changes in both migratory and resident DCs were also detected on day 14 in MLN of TNFRp55-/- mice. In vitro, LPS-stimulated migratory TNFRp55-/- DCs of MLN increased IL-12/23p40 compared with wild-type mice. In addition, TNFRp55-/- bone marrow-derived DCs in a TNFRp55-/- MLN microenvironment exhibited higher expression of CCR7 after Y. enterocolitica infection. The major intestinal DC subsets (CD103+CD11b-, CD103-CD11b+, and CD103+CD11b+) were found in the RLN of Y. enterocolitica-infected TNFRp55-/- mice. Fingolimod (FTY720) treatment of Y. enterocolitica-infected mice reduced the CD11b- subset of migratory DCs in RLN of TNFRp55-/- mice and significantly suppressed the severity of ReA in these mice. This result was associated with decreased articular IL-12/23p40 and IFN-γ levels. In vitro FTY720 treatment downregulated CCR7 on Y. enterocolitica-infected bone marrow-derived DCs and purified MLN DCs, which may explain the mechanism underlying the impairment of DCs in RLN induced by FTY720. Taken together, data indicate the migration of intestinal DCs to RLN and the contribution of these cells in the immunopathogenesis of ReA, which may provide evidence for controlling this disease.


Subject(s)
Arthritis, Reactive/immunology , Dendritic Cells/immunology , Lymph Nodes/immunology , Mesentery/immunology , Receptors, Tumor Necrosis Factor, Type I/metabolism , Tumor Necrosis Factor Decoy Receptors/metabolism , Yersinia Infections/immunology , Yersinia enterocolitica/immunology , Animals , Arthritis, Reactive/metabolism , Dendritic Cells/metabolism , Lymph Nodes/metabolism , Male , Mesentery/metabolism , Mice , Mice, Inbred C57BL , Prohibitins , Receptors, Tumor Necrosis Factor, Type I/immunology , Signal Transduction/immunology , Tumor Necrosis Factor Decoy Receptors/immunology , Yersinia Infections/metabolism
5.
Nat Immunol ; 10(1): 66-74, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19029905

ABSTRACT

The human body contains over 500 individual lymph nodes, yet the biology of their formation is poorly understood. Here we identify human lymphoid tissue-inducer cells (LTi cells) as lineage-negative RORC+ CD127+ cells with the functional ability to interact with mesenchymal cells through lymphotoxin and tumor necrosis factor. Human LTi cells were committed natural killer (NK) cell precursors that produced interleukin 17 (IL-17) and IL-22. In vitro, LTi cells gave rise to RORC+ CD127+ NK cells that retained the ability to produce IL-17 and IL-22. Postnatally, similar populations of LTi cell-like cells and RORC+ CD127+ NK cells were present in tonsils, and both secreted IL-17 and IL-22 but no interferon-gamma. Our data indicate that lymph node organogenesis is controlled by an NK cell precursor population with adaptive immune features and demonstrate a previously unappreciated link between the innate and adaptive immune systems.


Subject(s)
Interleukin-17/biosynthesis , Lymph Nodes/embryology , Lymph Nodes/immunology , Natural Killer T-Cells/immunology , Organogenesis , Precursor Cells, T-Lymphoid/immunology , Animals , CD56 Antigen/metabolism , Cell Differentiation , Cells, Cultured , Humans , Immunity, Cellular , Immunity, Innate , Interferon-gamma/biosynthesis , Interleukin-7 Receptor alpha Subunit/immunology , Interleukins/biosynthesis , Lymph Nodes/cytology , Lymphoid Tissue/embryology , Lymphoid Tissue/immunology , Lymphotoxin-alpha/immunology , Mesentery/embryology , Mesentery/immunology , Mice , Nuclear Receptor Subfamily 1, Group F, Member 3 , Palatine Tonsil/cytology , Palatine Tonsil/immunology , Receptors, Retinoic Acid/immunology , Receptors, Thyroid Hormone/immunology , Spleen/embryology , Spleen/immunology , Interleukin-22
6.
J Immunol ; 202(1): 260-267, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30487173

ABSTRACT

Salmonella infection is a globally important cause of gastroenteritis and systemic disease and is a useful tool to study immune responses in the intestine. Although mechanisms leading to immune responses against Salmonella have been extensively studied, questions remain about how bacteria travel from the intestinal mucosa to the mesenteric lymph nodes (MLN), a key site for Ag presentation. In this study, we used a mouse model of infection with Salmonella enterica serovar Typhimurium (STM) to identify changes in intestinal immune cells induced during early infection. We then used fluorescently labeled STM to identify interactions with immune cells from the site of infection through migration in lymph to the MLN. We show that viable STM can be carried in the lymph by any subset of migrating dendritic cells but not by macrophages. Moreover, approximately half of the STM in lymph are not associated with cells at all and travel autonomously. Within the MLN, STM associates with dendritic cells and B cells but predominantly with MLN-resident macrophages. In conclusion, we describe the routes used by STM to spread systemically in the period immediately postinfection. This deeper understanding of the infection process could open new avenues for controlling it.


Subject(s)
Dendritic Cells/immunology , Intestinal Mucosa/microbiology , Lymph Nodes/microbiology , Macrophages/immunology , Mesentery/immunology , Salmonella typhi/physiology , Typhoid Fever/immunology , Animals , Dendritic Cells/microbiology , Disease Models, Animal , Host-Pathogen Interactions , Humans , Intestinal Mucosa/immunology , Lymph Nodes/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Typhoid Fever/microbiology
7.
Am J Physiol Heart Circ Physiol ; 319(3): H705-H721, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32762560

ABSTRACT

Myeloperoxidase (MPO)-derived hypochlorous (HOCl) reacts with membrane plasmalogens to yield α-chlorofatty aldehydes such as 2-chlorofatty aldehyde (2-ClFALD) and its metabolite 2-chlorofatty acid (2-ClFA). Recent studies showed that 2-ClFALD and 2-ClFA serve as mediators of the inflammatory responses to sepsis by as yet unknown mechanisms. Since no scavenger for chlorinated lipids is available and on the basis of the well-established role of the MPO/HOCl/chlorinated lipid axis in inflammatory responses, we hypothesized that treatment with MPO inhibitors (N-acetyl lysyltyrosylcysteine amide or 4-aminobenzoic acid hydrazide) would inhibit inflammation and proinflammatory mediator expression induced by cecal ligation and puncture (CLP). We used intravital microscopy to quantify in vivo inflammatory responses in Sham and CLP rats with or without MPO inhibition. Small intestines, mesenteries, and lungs were collected to assess changes in MPO-positive staining and lung injury, respectively, as well as free 2-ClFA and proinflammatory mediators levels. CLP caused neutrophil infiltration, 2-ClFA generation, acute lung injury, leukocyte-/platelet-endothelium interactions, mast cell activation (MCA), plasminogen activator inhibitor-1 (PAI-1) production, and the expression of several cytokines, chemokines, and vascular endothelial growth factor, changes that were reduced by MPO inhibition. Pretreatment with a PAI-1 inhibitor or MC stabilizer prevented CLP-induced leukocyte-endothelium interactions and MCA, and abrogated exogenous 2-ClFALD-induced inflammatory responses. Thus, we provide evidence that MPO instigates these inflammatory changes in CLP and that chlorinated lipids may serve as a mechanistic link between the enzymatic activity of MPO and PAI-1- and mast cell-dependent adhesive interactions, providing a rationale for new therapeutic interventions in sepsis.NEW & NOTEWORTHY Using two distinct myeloperoxidase (MPO) inhibitors, we show for the first time that MPO plays an important role in producing increases in free 2-chlorofatty aldehyde (2-ClFALD)-a powerful proinflammatory chlorinated lipid in plasma and intestine-a number of cytokines and other inflammatory mediators, leukocyte and platelet rolling and adhesion in postcapillary venules, and lung injury in a cecal ligation and puncture model of sepsis. In addition, the use of a plasminogen activator inhibitor-1 (PAI-1) inhibitor or a mast cell stabilizer prevented inflammatory responses in CLP-induced sepsis. PAI-1 inhibition also prevented the proinflammatory responses to exogenous 2-ClFALD superfusion. Thus, our study provides some of the first evidence that MPO-derived free 2-ClFA plays an important role in CLP-induced sepsis by a PAI-1- and mast cell-dependent mechanism.


Subject(s)
Cecum/microbiology , Fatty Acids/metabolism , Hypochlorous Acid/metabolism , Inflammation Mediators/metabolism , Inflammation/enzymology , Peroxidase/metabolism , Sepsis/enzymology , Aldehydes/metabolism , Animals , Anti-Inflammatory Agents/pharmacology , Cecum/surgery , Cytokines/metabolism , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Inflammation/immunology , Inflammation/microbiology , Inflammation/prevention & control , Inflammation Mediators/antagonists & inhibitors , Intestine, Small/enzymology , Intestine, Small/immunology , Ligation , Lung/enzymology , Lung/immunology , Mast Cells/enzymology , Mast Cells/immunology , Mesentery/enzymology , Mesentery/immunology , Peroxidase/antagonists & inhibitors , Plasminogen Activator Inhibitor 1/metabolism , Punctures , Rats, Sprague-Dawley , Sepsis/immunology , Sepsis/microbiology , Sepsis/prevention & control , Signal Transduction
8.
Am J Pathol ; 189(2): 370-390, 2019 02.
Article in English | MEDLINE | ID: mdl-30660331

ABSTRACT

Commensal gut microbiota-host immune responses are experimentally delineated via gnotobiotic animal models or alternatively by antibiotic perturbation of gut microbiota. Osteoimmunology investigations in germ-free mice, revealing that gut microbiota immunomodulatory actions critically regulate physiologic skeletal development, highlight that antibiotic perturbation of gut microbiota may dysregulate normal osteoimmunological processes. We investigated the impact of antibiotic disruption of gut microbiota on osteoimmune response effects in postpubertal skeletal development. Sex-matched C57BL/6T mice were administered broad-spectrum antibiotics or vehicle-control from the age of 6 to 12 weeks. Antibiotic alterations in gut bacterial composition and skeletal morphology were sex dependent. Antibiotics did not influence osteoblastogenesis or endochondral bone formation, but notably enhanced osteoclastogenesis. Unchanged Tnf or Ccl3 expression in marrow and elevated tumor necrosis factor-α and chemokine (C-C motif) ligand 3 in serum indicated that the pro-osteoclastic effects of the antibiotics are driven by increased systemic inflammation. Antibiotic-induced broad changes in adaptive and innate immune cells in mesenteric lymph nodes and spleen demonstrated that the perturbation of gut microbiota drives a state of dysbiotic hyperimmune response at secondary lymphoid tissues draining local gut and systemic circulation. Antibiotics up-regulated the myeloid-derived suppressor cells, immature myeloid progenitor cells known for immunosuppressive properties in pathophysiologic inflammatory conditions. Myeloid-derived suppressor cell-mediated immunosuppression can be antigen specific. Therefore, antibiotic-induced broad suppression of major histocompatibility complex class II antigen presentation genes in bone marrow discerns that antibiotic perturbation of gut microbiota dysregulates critical osteoimmune cross talk.


Subject(s)
Anti-Bacterial Agents/adverse effects , Gastrointestinal Microbiome , Osteogenesis , Sexual Maturation , Animals , Anti-Bacterial Agents/pharmacology , Chemokine CCL3/immunology , Female , Gastrointestinal Microbiome/drug effects , Gastrointestinal Microbiome/immunology , Lymph Nodes/immunology , Lymph Nodes/pathology , Male , Mesentery/immunology , Mesentery/pathology , Mice , Myeloid-Derived Suppressor Cells/immunology , Myeloid-Derived Suppressor Cells/pathology , Osteoclasts/immunology , Osteoclasts/pathology , Osteogenesis/drug effects , Osteogenesis/immunology , Sexual Maturation/drug effects , Sexual Maturation/immunology , Spleen/immunology , Spleen/pathology , Tumor Necrosis Factor-alpha/immunology
9.
Blood ; 131(16): 1858-1869, 2018 04 19.
Article in English | MEDLINE | ID: mdl-29463561

ABSTRACT

Conditioning-induced damage of the intestinal tract plays a critical role during the onset of acute graft-versus-host disease (GVHD). Therapeutic interference with these early events of GVHD is difficult, and currently used immunosuppressive drugs mainly target donor T cells. However, not donor T cells but neutrophils reach the sites of tissue injury first, and therefore could be a potential target for GVHD prevention. A detailed analysis of neutrophil fate during acute GVHD and the effect on T cells is difficult because of the short lifespan of this cell type. By using a novel photoconverter reporter system, we show that neutrophils that had been photoconverted in the ileum postconditioning later migrated to mesenteric lymph nodes (mLN). This neutrophil migration was dependent on the intestinal microflora. In the mLN, neutrophils colocalized with T cells and presented antigen on major histocompatibility complex (MHC)-II, thereby affecting T cell expansion. Pharmacological JAK1/JAK2 inhibition reduced neutrophil influx into the mLN and MHC-II expression, thereby interfering with an early event in acute GVHD pathogenesis. In agreement with this finding, neutrophil depletion reduced acute GVHD. We conclude that neutrophils are attracted to the ileum, where the intestinal barrier is disrupted, and then migrate to the mLN, where they participate in alloantigen presentation. JAK1/JAK2-inhibition can interfere with this process, which provides a potential therapeutic strategy to prevent early events of tissue damage-related innate immune cell activation and, ultimately, GVHD.


Subject(s)
Cell Communication/immunology , Graft vs Host Disease/immunology , Ileum/immunology , Lymph Nodes/immunology , Mesentery/immunology , Neutrophils/immunology , Acute Disease , Animals , Cell Communication/drug effects , Cell Communication/genetics , Graft vs Host Disease/drug therapy , Graft vs Host Disease/genetics , Graft vs Host Disease/pathology , Ileum/pathology , Janus Kinase 1/antagonists & inhibitors , Janus Kinase 1/genetics , Janus Kinase 1/immunology , Janus Kinase 2/antagonists & inhibitors , Janus Kinase 2/genetics , Janus Kinase 2/immunology , Lymph Nodes/pathology , Mesentery/pathology , Mice , Mice, Inbred BALB C , Mice, Knockout , Neutrophil Infiltration/drug effects , Neutrophil Infiltration/genetics , Neutrophil Infiltration/immunology , Neutrophils/pathology , Protein Kinase Inhibitors/pharmacology
10.
Immunity ; 34(2): 247-57, 2011 Feb 25.
Article in English | MEDLINE | ID: mdl-21333555

ABSTRACT

Although both conventional dendritic cells (cDCs) and plasmacytoid dendritic cells (pDCs) are present in the gut-associated lymphoid tissues (GALT), the roles of pDCs in the gut remain largely unknown. Here we show a critical role for pDCs in T cell-independent (TI) IgA production by B cells in the GALT. When pDCs of the mesenteric lymph nodes (MLNs) and Peyer's patches (PPs) (which are representative GALT) were cultured with naive B cells to induce TI IgA class switch recombination (CSR), IgA production was substantially higher than in cocultures of these cells with cDCs. IgA production was dependent on APRIL and BAFF production by pDCs. Importantly, pDC expression of APRIL and BAFF was dependent on stromal cell-derived type I IFN signaling under steady-state conditions. Our findings provide insight into the molecular basis of pDC conditioning to induce mucosal TI IgA production, which may lead to improvements in vaccination strategies and treatment for mucosal-related disorders.


Subject(s)
Dendritic Cells/immunology , Immunity, Mucosal/immunology , Immunoglobulin A, Secretory/biosynthesis , Immunoglobulin Class Switching , Adoptive Transfer , Animals , B-Cell Activating Factor/physiology , Coculture Techniques , Cytokines/physiology , Germ-Free Life , Immunoglobulin A, Secretory/genetics , Interferon Type I/physiology , Lymph Nodes/immunology , Mesentery/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Peyer's Patches/immunology , Receptor Cross-Talk , Receptors, Antigen, T-Cell, alpha-beta/deficiency , Receptors, Antigen, T-Cell, alpha-beta/immunology , Receptors, Antigen, T-Cell, gamma-delta/deficiency , Receptors, Antigen, T-Cell, gamma-delta/immunology , Stromal Cells/immunology , Tumor Necrosis Factor Ligand Superfamily Member 13/physiology
11.
J Surg Res ; 256: 220-230, 2020 12.
Article in English | MEDLINE | ID: mdl-32711179

ABSTRACT

BACKGROUND: Immune dysfunction is associated with posthemorrhagic shock mesenteric lymph (PHSML) return. To determine the proliferation and cytokine production capacity of CD4+ T lymphocytes, the effect of PHSML drainage on spleen CD4+ T lymphocytes in a mouse model of hemorrhagic shock was assessed. METHODS: The normal spleen CD4+ T lymphocytes were in vitro incubated with either drained normal mesenteric lymph (NML), PHSML during hypotension (PHSML-H), or PHSML from 0 h to 3 h after resuscitation (PHSML-R) to verify direct proliferation effects of PHSML. RESULTS: Hemorrhagic shock led to reduction of proliferation and mRNA expression of interleukin 2 (IL-2) and IL-2 receptor in CD4+ T lymphocytes and to decrease in IL-2 and interferon γ (IFN-γ) levels in supernatants. In contrast, the interleukin-4 levels were increased. These effects were reversed by PHSML drainage. Moreover, NML incubation promoted CD4+ T lymphocyte proliferation, whereas both PHSML-H and PHSML-R treatment had a biphasic effects on CD4+ T lymphocyte proliferation, exhibiting an enhanced effect at early stages and an inhibitory effect at later stages. Compared with NML, PHSML-H increased IL-2 expression at 12 h, but decreased expression of both IL-2 and IFN-γ at 24 h. By contrast, PHSML-R induced significant increases in IL-2 and IFN-γ levels at 24 h. Interleukin-4 expression in CD4+ T lymphocytes was reduced at 12 h, but augmented at 24 h after incubation with either PHSML-H or PHSML-R. CONCLUSIONS: The results indicate that PHSML has a direct inhibitory effect on CD4+ T lymphocyte proliferation that induces an inflammatory response, which is associated with cellular immune dysfunction.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Lymph/immunology , Mesentery/immunology , Shock, Hemorrhagic/complications , Systemic Inflammatory Response Syndrome/immunology , Animals , Cell Proliferation , Cells, Cultured , Disease Models, Animal , Humans , Immunity, Cellular , Interferon-gamma/metabolism , Interleukin-2/metabolism , Interleukin-4/metabolism , Lymph/metabolism , Lymphatic Vessels , Lymphocyte Count , Male , Mesentery/metabolism , Mice , Primary Cell Culture , Receptors, Interleukin-2/metabolism , Shock, Hemorrhagic/blood , Shock, Hemorrhagic/immunology , Systemic Inflammatory Response Syndrome/blood
12.
Bull Exp Biol Med ; 168(4): 512-516, 2020 Feb.
Article in English | MEDLINE | ID: mdl-32147763

ABSTRACT

We performed a correlation analysis of the morphometric parameters of mesenteric lymph nodes and cytokine content in the lymph of thoracic duct in rats with chemically induced breast cancer. The study showed that activity of the local immune response in the lymph nodes in breast cancer is aimed at antitumor protection. In breast cancer, the area of the paracortical zone remained at the level of the intact group, while the area of lymphoid nodules with germinative centers and the area of medullary substance increased; the number of macrophages in the thymus-dependent zone and zone responsible for humoral immunity also increased. The following positive correlations were revealed: in germinative centers and medullary substance, number of mitotic cells correlated with cytokine IL-5 content and the number of medium lymphocytes correlated with the content of chemokine MIP-1α; in the germinative centers, the number of immunoblasts correlated with the level of cytokine GRO/KC, in the paracortical zone, the number of macrophages correlated with the level of chemokine MCP-1, the number of reticular cells correlated with IL-6 and M-CSF content; in medullary substance, the number of small lymphocytes and mature cells plasma cells (their content was reduced) correlated with the level of chemokine GRO/KC, which can be caused by their migration from the lymph node.


Subject(s)
Gene Expression Regulation, Neoplastic , Lymph Nodes/pathology , Mammary Glands, Animal/pathology , Mammary Neoplasms, Experimental/genetics , Mesentery/pathology , Thoracic Duct/pathology , Animals , Chemokine CCL2/genetics , Chemokine CCL2/immunology , Chemokine CCL3/genetics , Chemokine CCL3/immunology , Chemokine CXCL1/genetics , Chemokine CXCL1/immunology , Female , Interleukin-5/genetics , Interleukin-5/immunology , Interleukin-6/genetics , Interleukin-6/immunology , Lymph Nodes/immunology , Lymphatic Metastasis , Lymphocytes/immunology , Lymphocytes/pathology , Macrophage Colony-Stimulating Factor/genetics , Macrophage Colony-Stimulating Factor/immunology , Macrophages/immunology , Macrophages/pathology , Mammary Glands, Animal/drug effects , Mammary Glands, Animal/immunology , Mammary Neoplasms, Experimental/chemically induced , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/pathology , Mesentery/immunology , Methylnitrosourea/administration & dosage , Rats , Rats, Wistar , Thoracic Duct/immunology
13.
Cell Immunol ; 337: 33-41, 2019 03.
Article in English | MEDLINE | ID: mdl-30770094

ABSTRACT

Non-alcoholic fatty liver disease (NAFLD) is characterized by altered intestinal microbiota and intestinal immune disorder. Here we investigated the role of mesenteric lymph node (MLN) CD4+ T lymphocytes in NAFLD. In high fat diet (HFD)-fed mice, the percentage ratios of Th1 to Th2 cells and Th17 to Treg cells were imbalanced in MLNs. Co-culture assays showed MLN CD4+ T lymphocytes from HFD-fed mice tended to migrate to the liver and promoted hepatic inflammation. Adoptive transfer of MLN CD4+ T lymphocytes from NAFLD mice to HFD-fed mice resulted in higher transaminase, worse hepatic inflammation and lipid accumulation. Antibiotics and probiotics were administrated to regulate intestinal microbiota, and the restoration of MLN Th1/Th2 and Th17/Treg cells in alleviated NAFLD were found. In summary, MLNs CD4+ T subtype cells may involve in NAFLD, and the restoration of MLN CD4+ T subtype cells ratio by regulating intestinal bacteria could be the new strategies.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Mesentery/immunology , Non-alcoholic Fatty Liver Disease/immunology , Animals , Cell Movement/immunology , Cytokines , Diet, High-Fat , Inflammation/pathology , Liver/immunology , Lymph Nodes/immunology , Lymphocyte Count , Male , Mice , Mice, Inbred C57BL , Peritoneal Diseases/immunology , T-Lymphocytes, Helper-Inducer/immunology
14.
Nature ; 494(7435): 116-20, 2013 Feb 07.
Article in English | MEDLINE | ID: mdl-23334413

ABSTRACT

The intestinal microbiota has a critical role in immune system and metabolic homeostasis, but it must be tolerated by the host to avoid inflammatory responses that can damage the epithelial barrier separating the host from the luminal contents. Breakdown of this regulation and the resulting inappropriate immune response to commensals are thought to lead to the development of inflammatory bowel diseases such as Crohn's disease and ulcerative colitis. We proposed that the intestinal immune system is instructed by the microbiota to limit responses to luminal antigens. Here we demonstrate in mice that, at steady state, the microbiota inhibits the transport of both commensal and pathogenic bacteria from the lumen to a key immune inductive site, the mesenteric lymph nodes (MLNs). However, in the absence of Myd88 or under conditions of antibiotic-induced dysbiosis, non-invasive bacteria were trafficked to the MLNs in a CCR7-dependent manner, and induced both T-cell responses and IgA production. Trafficking was carried out by CX(3)CR1(hi) mononuclear phagocytes, an intestinal-cell population previously reported to be non-migratory. These findings define a central role for commensals in regulating the migration to the MLNs of CX(3)CR1(hi) mononuclear phagocytes endowed with the ability to capture luminal bacteria, thereby compartmentalizing the intestinal immune response to avoid inflammation.


Subject(s)
Immunity, Mucosal/immunology , Lymph Nodes/immunology , Lymph Nodes/microbiology , Mesentery/immunology , Metagenome/physiology , Phagocytes/metabolism , Receptors, Chemokine/metabolism , Animals , Anti-Bacterial Agents/pharmacology , Antigens, Bacterial/immunology , CX3C Chemokine Receptor 1 , Cell Movement , Dendritic Cells/cytology , Dendritic Cells/immunology , Immunity, Mucosal/drug effects , Immunoglobulin A/immunology , Inflammation/immunology , Intestinal Mucosa/cytology , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Metagenome/immunology , Mice , Mice, Inbred C57BL , Myeloid Differentiation Factor 88/deficiency , Myeloid Differentiation Factor 88/metabolism , Phagocytes/cytology , Phagocytes/immunology , Phagocytes/microbiology , Phagocytosis , Receptors, CCR7/deficiency , Receptors, CCR7/genetics , Receptors, CCR7/metabolism , Salmonella/cytology , Salmonella/drug effects , Salmonella/immunology , T-Lymphocytes/immunology
15.
Proc Natl Acad Sci U S A ; 113(30): 8502-7, 2016 07 26.
Article in English | MEDLINE | ID: mdl-27402748

ABSTRACT

Memory γδ T cells are important for the clearance of Listeria monocytogenes infection in the intestinal mucosa. However, the mechanisms by which memory γδ T cells provide protection against secondary oral infection are poorly understood. Here we used a recombinant strain of L. monocytogenes that efficiently invades the intestinal epithelium to show that Vγ4(+) memory γδ T cells represent a resident memory (Trm) population in the mesenteric lymph nodes (MLNs). The γδ Trm exhibited a remarkably static pattern of migration that radically changed following secondary oral L. monocytogenes infection. The γδ Trms produced IL-17A early after rechallenge and formed organized clusters with myeloid cells surrounding L. monocytogenes replication foci only after a secondary oral infection. Antibody blocking studies showed that in addition to IL-17A, the chemokine receptor C-X-C chemokine receptor 3 (CXCR3) is also important to enable the local redistribution of γδ Trm cells and myeloid cells specifically near the sites of L. monocytogenes replication within the MLN to restrict bacterial growth and spread. Our findings support a role for γδ Trms in orchestrating protective immune responses against intestinal pathogens.


Subject(s)
Immunity, Innate/immunology , Interleukin-17/immunology , Listeria monocytogenes/immunology , Listeriosis/immunology , Receptors, Antigen, T-Cell, gamma-delta/immunology , T-Lymphocyte Subsets/immunology , Animals , Cell Movement/immunology , Female , Immunologic Memory/immunology , Interleukin-17/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Listeria monocytogenes/physiology , Listeriosis/metabolism , Listeriosis/microbiology , Lymph Nodes/immunology , Mesentery/immunology , Mice, Inbred BALB C , Mice, Transgenic , Myeloid Cells/immunology , Receptors, Antigen, T-Cell, gamma-delta/genetics , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Receptors, CXCR3/immunology , Receptors, CXCR3/metabolism , T-Lymphocyte Subsets/metabolism
16.
Eur J Immunol ; 47(12): 2142-2152, 2017 12.
Article in English | MEDLINE | ID: mdl-28833065

ABSTRACT

Intestinal regulatory T cells (Tregs) are fundamental in peripheral tolerance toward commensals and food-borne antigens. Accordingly, gut-draining mesenteric lymph nodes (mLNs) represent a site of efficient peripheral de novo Treg induction when compared to skin-draining peripheral LNs (pLNs), and we had recently shown that LN stromal cells substantially contribute to this process. Here, we aimed to unravel the underlying molecular mechanisms and generated immortalized fibroblastic reticular cell lines (iFRCs) from mLNs and pLNs, allowing unlimited investigation of this rare stromal cell subset. In line with our previous findings, mLN-iFRCs showed a higher Treg-inducing capacity when compared to pLN-iFRCs. RNA-seq analysis focusing on secreted molecules revealed a more tolerogenic phenotype of mLN- as compared to pLN-iFRCs. Remarkably, mLN-iFRCs produced substantial numbers of microvesicles (MVs) that carried elevated levels of TGF-ß when compared to pLN-iFRC-derived MVs, and these novel players of intercellular communication were shown to be responsible for the tolerogenic properties of mLN-iFRCs. Thus, stromal cells originating from mLNs contribute to peripheral tolerance by fostering de novo Treg induction using TGF-ß-carrying MVs. This finding provides novel insights into the subcellular/molecular mechanisms of de novo Treg induction and might serve as promising tool for future therapeutic applications to treat inflammatory disorders.


Subject(s)
Extracellular Vesicles/immunology , Lymph Nodes/immunology , Stromal Cells/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Cell Line , Extracellular Vesicles/genetics , Extracellular Vesicles/ultrastructure , Forkhead Transcription Factors/immunology , Forkhead Transcription Factors/metabolism , Gene Expression Profiling/methods , Mesentery/immunology , Mice, Inbred BALB C , Mice, Knockout , Microscopy, Electron, Scanning , Microscopy, Electron, Transmission , Stromal Cells/metabolism , Stromal Cells/ultrastructure , T-Lymphocytes, Regulatory/metabolism , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/immunology
17.
Curr Opin Gastroenterol ; 34(4): 183-186, 2018 07.
Article in English | MEDLINE | ID: mdl-29846262

ABSTRACT

PURPOSE OF REVIEW: The occurrence of creeping fat wrapping segments of inflamed gut represents a characteristic yet incompletely understood hallmark of Crohn's disease. Over the last decade, numerous studies have provided a limited understanding of this feature. Still, deciphering the detailed mechanisms and the pathophysiologic relevance of the interplay between creeping fat, barrier function and intestinal inflammation will be the aim of future studies. RECENT FINDINGS: The last 18 months have substantially contributed to this field, starting with an elegant three-dimensional study revealing B cell aggregates around lymphatic vessels embedded in the mesenteric fat, thus bringing back the idea that Crohn's disease might represent a 'lymphatic disease'. Furthermore, studies on a cellular level elucidated the interplay of mesenteric adipocytes, immune cells and intestinal epithelial cells. Last, imaging studies provide evidence indicating that changes depicted by computed tomography within the mesenteric fat compartment rather than of the bowel wall are predictive for the presence of endoscopic lesions. This underlines the impact of mesenteric changes on Crohn's disease activity. SUMMARY: The findings of the last 18 months further contribute to solving the puzzle that will ultimately reveal the role of the mesenteric fat tissue in the control of intestinal immunity and inflammation.


Subject(s)
Adipose Tissue/immunology , Adipose Tissue/physiopathology , Crohn Disease/immunology , Crohn Disease/pathology , Adipokines/metabolism , Adipose Tissue/pathology , Animals , Biomarkers/metabolism , Crohn Disease/physiopathology , Humans , Inflammation/immunology , Inflammation/pathology , Mesentery/immunology , Mesentery/pathology , Mesentery/physiopathology
18.
Br J Nutr ; 119(5): 486-495, 2018 03.
Article in English | MEDLINE | ID: mdl-29508690

ABSTRACT

At birth, when immune responses are insufficient, there begins the development of the defence capability against pathogens. Leptin and adiponectin, adipokines that are present in breast milk, have been shown to play a role in the regulation of immune responses. We report here, for the first time, the influence of in vivo adipokine supplementation on the intestinal immune system in early life. Suckling Wistar rats were daily supplemented with leptin (0·7 µg/kg per d, n 36) or adiponectin (35 µg/kg per d, n 36) during the suckling period. The lymphocyte composition, proliferation and cytokine secretion from mesenteric lymph node lymphocytes (on days 14 and 21), as well as intestinal IgA and IgM concentration (day 21), were evaluated. At day 14, leptin supplementation significantly increased the TCRαß + cell proportion in mesenteric lymph nodes, in particular owing to an increase in the TCRαß + CD8+ cell population. Moreover, the leptin or adiponectin supplementation promoted the early development CD8+ cells, with adiponectin being the only adipokine capable of enhancing the lymphoproliferative ability at the end of the suckling period. Although leptin decreased intestinal IgA concentration, it had a trophic effect on the intestine in early life. Supplementation of both adipokines modulated the cytokine profile during (day 14) and at the end (day 21) of the suckling period. These results suggest that leptin and adiponectin during suckling play a role in the development of mucosal immunity in early life.


Subject(s)
Adiponectin/pharmacology , Animals, Suckling , Dietary Supplements , Intestines/drug effects , Leptin/pharmacology , Lymph Nodes/drug effects , Lymphocytes/metabolism , Animals , Animals, Newborn/growth & development , Animals, Newborn/immunology , Animals, Suckling/growth & development , Animals, Suckling/immunology , CD8 Antigens/metabolism , Immunity, Mucosal/drug effects , Immunoglobulin A/metabolism , Immunoglobulin M/metabolism , Intestinal Mucosa/drug effects , Intestines/immunology , Mesentery/immunology , Rats, Wistar , Receptors, Antigen, T-Cell, alpha-beta/metabolism
19.
Eur J Immunol ; 46(5): 1119-31, 2016 05.
Article in English | MEDLINE | ID: mdl-26887964

ABSTRACT

T cells of the small intestine, including Th17 cells, are critically involved in host protection from microbial infection, and also contribute to the pathogenesis of small bowel inflammatory disorders. Accumulating evidence suggests that mesenteric lymph nodes (MLNs) play important roles in gut-tropic T-cell generation, although it is still unclear if MLNs are involved in the pathogenesis of small intestine inflammation. To address this issue, we analyzed the roles of both MLNs and Peyer's patches (PPs) by evaluating MLN- or PP-deficient mice in an experimental model of small intestine inflammation, induced by CD3-specific mAb injection. Interestingly, MLNs, but not PPs, were essential for the pathogenesis of intestinal inflammation, in particular the accumulation and infiltration of CD4(+) T-cell populations, including Th17 cells, from the blood. In addition, CD4(+) T-cell accumulation was dependent on the function of the α4 ß7 integrin. Furthermore, MLN removal led to a significantly reduced number of peripheral α4 ß7 (+) CD4(+) effector memory T cells under normal conditions, suggesting that MLNs may play a role in maintaining the number of gut-tropic CD4(+) effector memory T cells circulating in the blood. Taken together, the present study highlights the important role of MLNs in contributing to the pathogenesis of small intestine inflammation.


Subject(s)
Enteritis/immunology , Intestine, Small/immunology , Lymph Nodes/immunology , Mesentery/immunology , Peyer's Patches/immunology , Th17 Cells/immunology , Animals , Antibodies, Monoclonal/administration & dosage , CD3 Complex/immunology , CD4-Positive T-Lymphocytes/immunology , Immunologic Memory , Inflammation/pathology , Integrins/immunology , Intestine, Small/pathology , Mice
20.
J Immunol ; 193(1): 431-8, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-24899505

ABSTRACT

IL-17-expressing CD4+ T lymphocytes (Th17 cells) naturally reside in the intestine where specific cytokines and microbiota, such as segmented filamentous bacteria (SFB), promote their differentiation. Intestinal Th17 cells are thought to initially differentiate in the GALT and/or mesenteric lymph nodes upon Ag encounter and subsequently home to the lamina propria (LP) where they mediate effector functions. However, whether GALT and/or mesenteric lymph nodes are required for intestinal Th17 differentiation as well as how microbiota containing SFB regulate Ag-specific intestinal Th17 cells remain poorly defined. In this study, we observed that naive CD4+ T cells were abundant in the intestinal LP prior to weaning and that the accumulation of Th17 cells in response to microbiota containing SFB occurred in the absence of lymphotoxin-dependent lymphoid structures and the spleen. Furthermore, the differentiation of intestinal Th17 cells in the presence of microbiota containing SFB was dependent on MHC class II expression by CD11c+ cells. Lastly, the differentiation of Ag-specific Th17 cells required both the presence of cognate Ag and microbiota containing SFB. These findings suggest that microbiota containing SFB create an intestinal milieu that may induce Ag-specific Th17 differentiation against food and/or bacterial Ags directly in the intestinal LP.


Subject(s)
Bacteria/immunology , Cell Differentiation/immunology , Histocompatibility Antigens Class II/immunology , Intestines , Lymph Nodes/immunology , Mesentery/immunology , Th17 Cells/immunology , Animals , Antigens, Bacterial/immunology , Cell Differentiation/genetics , Histocompatibility Antigens Class II/genetics , Intestines/immunology , Intestines/microbiology , Mice , Mice, Knockout , Th17 Cells/cytology
SELECTION OF CITATIONS
SEARCH DETAIL