Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
J Leukoc Biol ; 2024 Mar 14.
Article in English | MEDLINE | ID: mdl-38484149

ABSTRACT

In chronic inflammation regulatory immune cells, such as regulatory T cells and myeloid-derived suppressor cells (MDCS) can develop. Local signals in the inflamed tissue, such as cytokines and eicosanoids, but also contact dependent signals, can promote MDSC development. In the liver, hepatic stellate cells (HSC) may provide such signals via the expression of CD44. MDSC generated in the presence of HSC and anti-CD44 antibodies were functionally and phenotypically analyzed. We found that both monocytic (M-) and polymorphonuclear (PMN-) MDSC generated in the presence of αCD44 antibodies were less suppressive towards T cells as measured by T cell proliferation and cytokine production. Moreover, both M- and PMN- MDSC were phenotypically altered. M-MDSC mainly changed their expression of CD80 and CD39, PMN-MDSC showed altered expression of CD80/86, PD-L1 and CCR2. Moreover, both PMN- and M-MDSC lost expression of Nos2 mRNA, whereas M-MDSC showed reduced expression of TGFb mRNA and PMN-MDSC reduced expression of Il10 mRNA. In summary, the presence of CD44 on hepatic stellate cells promotes the induction of both M- and PMN-MDSC, although the mechanisms by which these MDSC may increase suppressive function due to interaction with CD44 is only partially overlapping.

3.
Kidney Int ; 104(1): 74-89, 2023 07.
Article in English | MEDLINE | ID: mdl-36924892

ABSTRACT

Previous studies have identified a unique Treg population, which expresses the Th17 characteristic transcription factor RORγt. These RORγt+ Tregs possess enhanced immunosuppressive capacity, which endows them with great therapeutic potential. However, as a caveat, they are also capable of secreting pro-inflammatory IL-17A. Since the sum function of RORγt+ Tregs in glomerulonephritis (GN) remains unknown, we studied the effects of their absence. Purified CD4+ T cell populations, containing or lacking RORγt+ Tregs, were transferred into immunocompromised RAG1 knockout mice and the nephrotoxic nephritis model of GN was induced. Absence of RORγt+ Tregs significantly aggravated kidney injury, demonstrating overall kidney-protective properties. Analyses of immune responses showed that RORγt+ Tregs were broadly immunosuppressive with no preference for a particular type of T cell response. Further characterization revealed a distinct functional and transcriptional profile, including enhanced production of IL-10. Expression of the chemokine receptor CCR6 marked a particularly potent subset, whose absence significantly worsened GN. As an underlying mechanism, we found that chemokine CCL20 acting through receptor CCR6 signaling mediated expansion and activation of RORγt+ Tregs. Finally, we also detected an increase of CCR6+ Tregs in kidney biopsies, as well as enhanced secretion of chemokine CCL20 in 21 patients with anti-neutrophil cytoplasmic antibody associated GN compared to that of 31 healthy living donors, indicating clinical relevance. Thus, our data characterize RORγt+ Tregs as anti-inflammatory mediators of GN and identify them as promising target for Treg directed therapies.


Subject(s)
Glomerulonephritis , Nuclear Receptor Subfamily 1, Group F, Member 3 , Mice , Animals , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , T-Lymphocytes, Regulatory , Chemokine CCL20/genetics , Chemokine CCL20/metabolism , Kidney/pathology , Mice, Knockout , Th17 Cells , Receptors, CCR6/genetics , Receptors, CCR6/metabolism
4.
Cells ; 11(17)2022 08 25.
Article in English | MEDLINE | ID: mdl-36078047

ABSTRACT

Interleukin-2 is central to the induction and maintenance of both natural (nTreg) and induced Foxp3-expressing regulatory T cells (iTreg). Thus, signals that modulate IL-2 availability may, in turn, also influence Treg homeostasis. Using global knockout and cell-specific knockout mouse models, we evaluated the role of the small GTPase ADP-ribosylation factor 4d (Arl4d) in regulatory T-cell biology. We show that the expression of Arl4d in T cells restricts both IL-2 production and responsiveness to IL-2, as measured by the phosphorylation of STAT5. Arl4d-deficient CD4 T cells converted more efficiently into Foxp3+ iTreg in vitro in the presence of αCD3ε and TGFß, which was associated with their enhanced IL-2 secretion. As such, Arl4d-/- CD4 T cells induced significantly less colonic inflammation and lymphocytic infiltration in a model of transfer colitis. Thus, our data reveal a negative regulatory role for Arl4d in CD4 T-cell biology, limiting iTreg conversion via the restriction of IL-2 production, leading to reduced induction of Treg from conventional CD4 T cells.


Subject(s)
Interleukin-2 , T-Lymphocytes, Regulatory , ADP-Ribosylation Factors/metabolism , Animals , Forkhead Transcription Factors/metabolism , Interleukin-2/metabolism , Mice , Mice, Knockout , T-Lymphocytes, Regulatory/metabolism , Transforming Growth Factor beta/metabolism
5.
J Autoimmun ; 129: 102829, 2022 05.
Article in English | MEDLINE | ID: mdl-35468361

ABSTRACT

Systemic lupus erythematosus (SLE) is a common autoimmune disorder with a complex and poorly understood immuno-pathogenesis. Lupus nephritis (LN) is a frequent and difficult to treat complication, which causes high morbidity and mortality. The multifunctional cytokine amphiregulin (AREG) has been implicated in SLE pathogenesis, but its function in LN currently remains unknown. We thus studied the model of pristane-induced LN and found increasing renal and systemic AREG expression during the course of disease. Importantly, renal injury was significantly aggravated in the absence of AREG, revealing a net anti-inflammatory role. Analyses of immune responses showed dual effects. On the one hand, AREG enhanced activation of pro-inflammatory myeloid cells, which however did not play a major role for the course of LN. More importantly, on the other hand, AREG strongly suppressed pathogenic cytokine production by T helper effector cells. This effect was more general in nature and could be reproduced in response to antigen immunization. Since AREG has been postulated to downregulate T cell responses via enhancing Treg suppressive capacity, we followed up on this aspect. Interestingly, however, in vitro studies revealed potential direct and Treg independent effects of AREG on T helper effector cells. In favor of this notion, we found significantly enhanced T cell responses and consecutive aggravation of LN, only if epidermal growth factor receptor (EGFR) signaling was abrogated in total T cells, but not if the EGFR was absent on Tregs alone. Finally, we also found enhanced AREG expression in plasma and renal biopsies of patients with LN, supporting the relevance of our findings for human disease. In summary, our data identify AREG as an anti-inflammatory mediator of LN via broad downregulation of pathogenic T cell immunity. These findings further highlight the AREG/EGFR axis as a potential therapeutic target.


Subject(s)
Lupus Erythematosus, Systemic , Lupus Nephritis , Amphiregulin/genetics , Amphiregulin/metabolism , Amphiregulin/therapeutic use , Cytokines/metabolism , Down-Regulation , ErbB Receptors/metabolism , ErbB Receptors/therapeutic use , Humans , Lupus Erythematosus, Systemic/pathology , Lupus Nephritis/metabolism , T-Lymphocytes, Helper-Inducer/metabolism
6.
Int J Mol Sci ; 22(21)2021 Oct 27.
Article in English | MEDLINE | ID: mdl-34769006

ABSTRACT

Myeloid cells play an essential role in the maintenance of liver homeostasis, as well as the initiation and termination of innate and adaptive immune responses. In chronic hepatic inflammation, the production of transforming growth factor beta (TGF-ß) is pivotal for scarring and fibrosis induction and progression. TGF-ß signalling is tightly regulated via the Smad protein family. Smad7 acts as an inhibitor of the TGF-ß-signalling pathway, rendering cells that express high levels of it resistant to TGF-ß-dependent signal transduction. In hepatocytes, the absence of Smad7 promotes liver fibrosis. Here, we examine whether Smad7 expression in myeloid cells affects the extent of liver inflammation, injury and fibrosis induction during chronic liver inflammation. Using the well-established model of chronic carbon tetrachloride (CCl4)-mediated liver injury, we investigated the role of Smad7 in myeloid cells in LysM-Cre Smadfl/fl mice that harbour a myeloid-specific knock-down of Smad7. We found that the chronic application of CCl4 induces severe liver injury, with elevated serum alanine transaminase (ALT)/aspartate transaminase (AST) levels, centrilobular and periportal necrosis and immune-cell infiltration. However, the myeloid-specific knock-down of Smad7 did not influence these and other parameters in the CCl4-treated animals. In summary, our results suggest that, during long-term application of CCl4, Smad7 expression in myeloid cells and its potential effects on the TGF-ß-signalling pathway are dispensable for regulating the extent of chronic liver injury and inflammation.


Subject(s)
Carbon Tetrachloride/pharmacology , Inflammation/metabolism , Liver Cirrhosis/chemically induced , Liver Cirrhosis/metabolism , Liver Diseases/metabolism , Myeloid Cells/metabolism , Smad7 Protein/deficiency , Alanine Transaminase/metabolism , Animals , Disease Models, Animal , Hepatocytes/metabolism , Liver/metabolism , Male , Mice , Signal Transduction/physiology , Smad7 Protein/metabolism , Transforming Growth Factor beta/metabolism
7.
J Am Soc Nephrol ; 31(9): 1996-2012, 2020 09.
Article in English | MEDLINE | ID: mdl-32616537

ABSTRACT

BACKGROUND: Recent studies have identified the EGF receptor (EGFR) ligand amphiregulin (AREG) as an important mediator of inflammatory diseases. Both pro- and anti-inflammatory functions have been described, but the role of AREG in GN remains unknown. METHODS: The nephrotoxic nephritis model of GN was studied in AREG-/- mice after bone marrow transplantation, and in mice with myeloid cell-specific EGFR deficiency. Therapeutic utility of AREG neutralization was assessed. Furthermore, AREG's effects on renal cells and monocytes/macrophages (M/M) were analyzed. Finally, we evaluated AREG expression in human renal biopsies. RESULTS: Renal AREG mRNA was strongly upregulated in murine GN. Renal resident cells were the most functionally relevant source of AREG. Importantly, the observation that knockout mice showed significant amelioration of disease indicates that AREG is pathogenic in GN. AREG enhanced myeloid cell responses via inducing chemokine and colony stimulating factor 2 (CSF2) expression in kidney resident cells. Furthermore, AREG directly skewed M/M to a proinflammatory M1 phenotype and protected them from apoptosis. Consequently, anti-AREG antibody treatment dose-dependently ameliorated GN. Notably, selective abrogation of EGFR signaling in myeloid cells was sufficient to protect against nephritis. Finally, strong upregulation of AREG expression was also detected in kidneys of patients with two forms of crescentic GN. CONCLUSIONS: AREG is a proinflammatory mediator of GN via (1) enhancing renal pathogenic myeloid cell infiltration and (2) direct effects on M/M polarization, proliferation, and cytokine secretion. The AREG/EGFR axis is a potential therapeutic target for acute GN.


Subject(s)
Amphiregulin/physiology , Glomerulonephritis/etiology , Myeloid Cells/physiology , Animals , Cell Movement , Cells, Cultured , Chemokines/biosynthesis , ErbB Receptors/physiology , Glomerulonephritis/pathology , Granulocyte-Macrophage Colony-Stimulating Factor/biosynthesis , Humans , Macrophages/physiology , Mice , Mice, Inbred C57BL
8.
J Am Soc Nephrol ; 30(8): 1439-1453, 2019 08.
Article in English | MEDLINE | ID: mdl-31311828

ABSTRACT

BACKGROUND: New therapies blocking the IL-6 receptor (IL-6R) have recently become available and are successfully being used to treat inflammatory diseases like arthritis. Whether IL-6 blockers may help patients with kidney inflammation currently remains unknown. METHODS: To learn more about the complex role of CD4+ T cell-intrinsic IL-6R signaling, we induced nephrotoxic nephritis, a mouse model for crescentic GN, in mice lacking T cell-specific IL-6Ra. We used adoptive transfer experiments and studies in reporter mice to analyze immune responses and Treg subpopulations. RESULTS: Lack of IL-6Ra signaling in mouse CD4+ T cells impaired the generation of proinflammatory Th17 cells, but surprisingly did not ameliorate the course of GN. In contrast, renal damage was significantly reduced by restricting IL-6Ra deficiency to T effector cells and excluding Tregs. Detailed studies of Tregs revealed unaltered IL-10 production despite IL-6Ra deficiency. However, in vivo and in vitro, IL-6Ra classic signaling induced RORγt+Foxp3+ double-positive Tregs (biTregs), which carry the trafficking receptor CCR6 and have potent immunoregulatory properties. Indeed, lack of IL-6Ra significantly reduced Treg in vitro suppressive capacity. Finally, adoptive transfer of T cells containing IL-6Ra-/- Tregs resulted in severe aggravation of GN in mice. CONCLUSIONS: Our data refine the old paradigm, that IL-6 enhances Th17 responses and suppresses Tregs. We here provide evidence that T cell-intrinsic IL-6Ra classic signaling indeed induces the generation of Th17 cells but at the same time highly immunosuppressive RORγt+ biTregs. These results advocate caution and indicate that IL-6-directed therapies for GN need to be cell-type specific.


Subject(s)
Forkhead Transcription Factors/metabolism , Interleukin-6 Receptor alpha Subunit/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Receptors, Interleukin-6/metabolism , Signal Transduction , T-Lymphocytes, Regulatory/immunology , Animals , Cell Line , Crosses, Genetic , Female , Glomerulonephritis/metabolism , Glomerulonephritis/pathology , Immunosuppressive Agents/therapeutic use , Inflammation , Interleukin-6 Receptor alpha Subunit/genetics , Male , Mice , Mice, Inbred C57BL , Necrosis , Receptors, Interleukin-6/genetics , Th17 Cells/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...