Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
Add more filters










Publication year range
1.
Cell Commun Signal ; 22(1): 281, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38773618

ABSTRACT

BACKGROUND: Restoring impaired peripheral immune tolerance is the primary challenge in treating autoimmune diseases. Our previous research demonstrated the effectiveness of small spleen peptides (SSPs), a fraction of low molecular weight proteins, in inhibiting the progression of psoriatic arthritis, even in the presence of high levels of the proinflammatory cytokine TNFα in the bloodstream. When specifically targeting dendritic cells (DCs), SSPs transform them into tolerogenic cells, which efficiently induce the development of regulatory Foxp3+ Treg cells. In this study, we provide further insights into the mechanism of action of SSPs. RESULTS: We found that SSPs stimulate the activation of the mTOR signaling pathway in dendritic cells, albeit in a different manner than the classical immunogenic stimulus LPS. While LPS-induced activation is rapid, strong, and sustained, the activity induced by SSPs is delayed, less intense, yet still significant. These distinct patterns of activation, as measured by phosphorylation of key components of the pathway are also observed in response to other immunogenic and tolerogenic stimuli such as GM-CSF + IL-4 or IL-10 and TGFß. The disparity in mTOR activation between immunogenic and tolerogenic stimuli is quantitative rather than qualitative. In both cases, mTOR activation primarily occurs through the PI3K/Akt signaling axis and involves ERK and GSK3ß kinases, with minimal involvement of AMPK or NF-kB pathways. Furthermore, in the case of SSPs, mTOR activation seems to involve adenosine receptors. Additionally, we observed that DCs treated with SSPs exhibit an energy metabolism with high plasticity, which is typical of tolerogenic cells rather than immunogenic cells. CONCLUSION: Hence, the decision whether dendritic cells enter an inflammatory or tolerogenic state seems to rely on varying activation thresholds and kinetics of the mTOR signaling pathway.


Subject(s)
Dendritic Cells , Immune Tolerance , Signal Transduction , TOR Serine-Threonine Kinases , Dendritic Cells/immunology , Dendritic Cells/metabolism , Dendritic Cells/drug effects , TOR Serine-Threonine Kinases/metabolism , Signal Transduction/drug effects , Animals , Mice , Inflammation/metabolism , Kinetics , Lipopolysaccharides/pharmacology
2.
Biomolecules ; 14(4)2024 Apr 11.
Article in English | MEDLINE | ID: mdl-38672485

ABSTRACT

Restoring peripheral immune tolerance is crucial for addressing autoimmune diseases. An ancient mechanism in maintaining the balance between inflammation and tolerance is the ratio of extracellular ATP (exATP) and adenosine. Our previous research demonstrated the effectiveness of small spleen peptides (SSPs) in inhibiting psoriatic arthritis progression, even in the presence of the pro-inflammatory cytokine TNFα, by transforming dendritic cells (DCs) into tolerogenic cells and fostering regulatory Foxp3+ Treg cells. Here, we identified thymosins as the primary constituents of SSPs, but recombinant thymosin peptides were less efficient in inhibiting arthritis than SSPs. Since Tß4 is an ecto-ATPase-binding protein, we hypothesized that SSPs regulate exATP profiles. Real-time investigation of exATP levels in DCs revealed that tolerogenic stimulation led to robust de novo exATP synthesis followed by significant degradation, while immunogenic stimulation resulted in a less pronounced increase in exATP and less effective degradation. These contrasting exATP profiles were crucial in determining whether DCs entered an inflammatory or tolerogenic state, highlighting the significance of SSPs as natural regulators of peripheral immunological tolerance, with potential therapeutic benefits for autoimmune diseases. Finally, we demonstrated that the tolerogenic phenotype of SSPs is mainly influenced by adenosine receptors, and in vivo administration of SSPs inhibits psoriatic skin inflammation.


Subject(s)
Adenosine Triphosphate , Cell Differentiation , Dendritic Cells , Spleen , Dendritic Cells/metabolism , Dendritic Cells/drug effects , Dendritic Cells/immunology , Adenosine Triphosphate/metabolism , Adenosine Triphosphate/pharmacology , Animals , Cell Differentiation/drug effects , Spleen/cytology , Spleen/metabolism , Spleen/drug effects , Spleen/immunology , Mice , Thymosin/pharmacology , Thymosin/metabolism , Peptides/pharmacology , Arthritis, Psoriatic/drug therapy , Arthritis, Psoriatic/metabolism , Arthritis, Psoriatic/immunology , Humans , Mice, Inbred C57BL , Immune Tolerance/drug effects
3.
Mol Ther Oncolytics ; 31: 100741, 2023 Dec 19.
Article in English | MEDLINE | ID: mdl-38020062

ABSTRACT

Low pathogenic influenza A viruses (IAVs) have shown promising oncolytic potential in lung cancer-bearing mice. However, as replication-competent pathogens, they may cause side effects in immunocompromised cancer patients. To circumvent this problem, we genetically engineered nonreplicating IAVs lacking the hemagglutinin (HA) gene (ΔHA IAVs), but reconstituted the viral envelope with recombinant HA proteins to allow a single infection cycle. To optimize the therapeutic potential and improve immunomodulatory properties, these replication-incompetent IAVs were complemented with a murine interferon-gamma (mIFN-γ) gene. After intratracheal administration to transgenic mice that develop non-small cell lung cancer (NSCLC), the ΔHA IAVs induced potent tumor destruction. However, ΔHA IAVs armed with mIFN-γ exhibited an even stronger and more sustained effect, achieving 85% tumor reduction at day 12 postinfection. In addition, ΔHA-mIFN-γ viruses were proven to be efficient in recruiting and activating natural killer cells and macrophages from the periphery and in inducing cytotoxic T lymphocytes. Most important, both viruses, and particularly IFN-γ-encoding viruses, activated tumor-associated alveolar macrophages toward a proinflammatory M1-like phenotype. Therefore, replication-incompetent ΔHA-mIFN-γ-IAVs are safe and efficient oncolytic viruses that additionally exhibit immune cell activating properties and thus represent a promising innovative therapeutic option in the fight against NSCLC.

4.
FEBS J ; 289(15): 4622-4645, 2022 08.
Article in English | MEDLINE | ID: mdl-35176204

ABSTRACT

Four-and-a-half LIM domains protein 2 (FHL2) is an anti-hypertrophic adaptor protein that regulates cardiac myocyte signalling and function. Herein, we identified cardiomyopathy-associated 5 (CMYA5) as a novel FHL2 interaction partner in cardiac myocytes. In vitro pull-down assays demonstrated interaction between FHL2 and the N- and C-terminal regions of CMYA5. The interaction was verified in adult cardiac myocytes by proximity ligation assays. Immunofluorescence and confocal microscopy demonstrated co-localisation in the same subcellular compartment. The binding interface between FHL2 and CMYA5 was mapped by peptide arrays. Exposure of neonatal rat ventricular myocytes to a CMYA5 peptide covering one of the FHL2 interaction sites led to an increase in cell area at baseline, but a blunted response to chronic phenylephrine treatment. In contrast to wild-type hearts, loss or reduced FHL2 expression in Fhl2-targeted knockout mouse hearts or in a humanised mouse model of hypertrophic cardiomyopathy led to redistribution of CMYA5 into the perinuclear and intercalated disc region. Taken together, our results indicate a direct interaction of the two adaptor proteins FHL2 and CMYA5 in cardiac myocytes, which might impact subcellular compartmentation of CMYA5.


Subject(s)
Cardiomyopathy, Hypertrophic , Intracellular Signaling Peptides and Proteins , LIM-Homeodomain Proteins , Muscle Proteins , Myocytes, Cardiac , Transcription Factors , Animals , Cardiomyopathy, Hypertrophic/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , LIM-Homeodomain Proteins/genetics , LIM-Homeodomain Proteins/metabolism , Mice , Mice, Knockout , Muscle Proteins/genetics , Muscle Proteins/metabolism , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Rats , Signal Transduction , Transcription Factors/genetics , Transcription Factors/metabolism
5.
Mol Ther ; 30(2): 745-762, 2022 02 02.
Article in English | MEDLINE | ID: mdl-34450252

ABSTRACT

The major challenge in the treatment of autoimmune diseases is the restoration of the impaired peripheral immune tolerance that always accompanies the development of such diseases. Here, we show that small splenic peptides (SSPs) of whole spleen extract efficiently suppress the development of psoriatic arthritis in vivo, even in the presence of sustained levels of pro-inflammatory cytokines. SSPs target dendritic cells (DCs) and convert them into tolerogenic cells, which in turn differentiate naive CD4+ cells into Foxp3-expressing T regulatory cells (Tregs). The latter requires direct contact between SSP-activated DCs and naive CD4+ T cells via PD-1 and CTLA4 immune checkpoint receptors of T cells. Finally, depletion of Foxp3+ Tregs in vivo abrogated the protective effect of SSPs on psoriatic arthritis development. We hypothesize that SSPs represent an intrinsic component of the adaptive immune system responsible for the physiological maintenance of peripheral tolerance and that therapeutically administered SSPs are able to restore imbalanced peripheral tolerance in autoimmune diseases.


Subject(s)
Arthritis, Psoriatic , Immune Tolerance , Arthritis, Psoriatic/therapy , Cytokines , Dendritic Cells , Humans , Peripheral Tolerance , Spleen , T-Lymphocytes, Regulatory
6.
J Virol ; 95(20): e0067221, 2021 09 27.
Article in English | MEDLINE | ID: mdl-34319782

ABSTRACT

Influenza A virus (IAV) is the causative agent of flu disease that results in annual epidemics and occasional pandemics. IAV alters several signaling pathways of the cellular host response in order to promote its replication. Therefore, some of these pathways can serve as targets for novel antiviral agents. Here, we show that c-Jun NH2-terminal kinase (JNK)-interacting protein 4 (JIP4) is dynamically phosphorylated in IAV infection. The lack of JIP4 resulted in higher virus titers, with significant differences in viral protein and mRNA accumulation as early as within the first replication cycle. In accordance, decreased IAV titers and protein accumulation were observed during the overexpression of JIP4. Strikingly, the antiviral function of JIP4 does not originate from modulation of JNK or p38 mitogen-activated protein kinase (MAPK) pathways or from altered expression of interferons or interferon-stimulated genes but rather originates from a direct reduction of viral polymerase activity. Furthermore, the interference of JIP4 with IAV replication seems to be linked to the phosphorylation of the serine at position 730 that is sufficient to impede the viral polymerase. Collectively, we provide evidence that JIP4, a host protein modulated in IAV infection, exhibits antiviral properties that are dynamically controlled by its phosphorylation at S730. IMPORTANCE Influenza A virus (IAV) infection is a world health concern, and current treatment options encounter high rates of resistance. Our group investigates host pathways modified in IAV infection as promising new targets. The host protein JIP4 is dynamically phosphorylated in IAV infection. JIP4 absence resulted in higher virus titers and viral protein and mRNA accumulation within the first replication cycle. Accordingly, decreased IAV titers and protein accumulation were observed during JIP4 overexpression. Strikingly, the antiviral function of JIP4 does not originate from modulation of JNK or p38 MAPK pathways or from altered expression of interferons or interferon-stimulated genes but rather originates from a reduction in viral polymerase activity. The interference of JIP4 with IAV replication is linked to the phosphorylation of serine 730. We provide evidence that JIP4, a host protein modulated in IAV infection, exhibits antiviral properties that are dynamically controlled by its phosphorylation at S730.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Influenza A virus/metabolism , A549 Cells , Animals , Chlorocebus aethiops , Dogs , Host-Pathogen Interactions/genetics , Humans , Immune Evasion/genetics , Immunity, Innate/genetics , Influenza A virus/physiology , Influenza, Human/virology , Interferons/genetics , Madin Darby Canine Kidney Cells , Phosphorylation , Signal Transduction/genetics , Vero Cells , Virus Replication/genetics , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
7.
Immunity ; 54(5): 1002-1021.e10, 2021 05 11.
Article in English | MEDLINE | ID: mdl-33761330

ABSTRACT

Arthritis typically involves recurrence and progressive worsening at specific predilection sites, but the checkpoints between remission and persistence remain unknown. Here, we defined the molecular and cellular mechanisms of this inflammation-mediated tissue priming. Re-exposure to inflammatory stimuli caused aggravated arthritis in rodent models. Tissue priming developed locally and independently of adaptive immunity. Repeatedly stimulated primed synovial fibroblasts (SFs) exhibited enhanced metabolic activity inducing functional changes with intensified migration, invasiveness and osteoclastogenesis. Meanwhile, human SF from patients with established arthritis displayed a similar primed phenotype. Transcriptomic and epigenomic analyses as well as genetic and pharmacological targeting demonstrated that inflammatory tissue priming relies on intracellular complement C3- and C3a receptor-activation and downstream mammalian target of rapamycin- and hypoxia-inducible factor 1α-mediated metabolic SF invigoration that prevents activation-induced senescence, enhances NLRP3 inflammasome activity, and in consequence sensitizes tissue for inflammation. Our study suggests possibilities for therapeutic intervention abrogating tissue priming without immunosuppression.


Subject(s)
Complement System Proteins/immunology , Fibroblasts/immunology , Inflammation/immunology , Synovial Membrane/immunology , Adaptive Immunity/immunology , Animals , Arthritis, Rheumatoid/immunology , Cell Line , Dogs , Humans , Inflammation Mediators/immunology , Madin Darby Canine Kidney Cells , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Rats, Wistar , Signal Transduction/immunology
8.
Oncoimmunology ; 10(1): 1885778, 2021 02 17.
Article in English | MEDLINE | ID: mdl-33643696

ABSTRACT

Non-small cell lung cancers (NSCLCs) establish a highly immunosuppressive tumor microenvironment supporting cancer growth. To interfere with cancer-mediated immunosuppression, selective immune-checkpoint inhibitors (ICIs) have been approved as a standard-of-care treatment for NSCLCs. However, the majority of patients poorly respond to ICI-based immunotherapies. Oncolytic viruses are amongst the many promising immunomodulatory treatments tested as standalone therapy or in combination with ICIs to improve therapeutic outcome. Previously, we demonstrated the oncolytic and immunomodulatory efficacy of low-pathogenic influenza Aviruses (IAVs) against NSCLCs in immunocompetent transgenic mice with alung-specific overexpression of active Raf kinase (Raf-BxB). IAV infection not only resulted in significant primary virus-induced oncolysis, but also caused afunctional reversion of tumor-associated macrophages (TAMs) comprising additional anti-cancer activity. Here we show that NSCLCs as well as TAMs and cytotoxic immune cells overexpress IC molecules of the PD-L2/PD-1 and B7-H3 signaling axes. Thus, we aimed to combine oncolytic IAV-infection with ICIs to exploit the benefits of both anti-cancer approaches. Strikingly, IAV infection combined with the novel B7-H3 ICI led to increased levels of M1-polarized alveolar macrophages and increased lung infiltration by cytotoxic Tlymphocytes, which finally resulted in significantly improved oncolysis of about 80% of existing tumors. In contrast, application of clinically approved α-PD-1 IC antibodies alone or in combination with oncolytic IAV did not provide additional oncolytic or immunomodulatory efficacy. Thus, individualized therapy with synergistically acting oncolytic IAV and B7-H3 ICI might be an innovative future approach to target NSCLCs that are resistant to approved ICIs in patients.


Subject(s)
Influenza, Human , Lung Neoplasms , Oncolytic Viruses , Orthomyxoviridae , Animals , Humans , Immune Checkpoint Inhibitors , Lung , Lung Neoplasms/therapy , Mice , Oncolytic Viruses/genetics , Tumor Microenvironment
9.
J Virol ; 95(6)2021 02 24.
Article in English | MEDLINE | ID: mdl-33408177

ABSTRACT

Influenza A virus (IAV) nonstructural protein 1 (NS1) is a protein with multiple functions that are regulated by phosphorylation. Phosphoproteomic screening of H1N1 virus-infected cells revealed that NS1 was phosphorylated at serine 205 in intermediate stages of the viral life cycle. Interestingly, S205 is one of six amino acid changes in NS1 of post-pandemic H1N1 viruses currently circulating in humans compared to the original swine-origin 2009 pandemic (H1N1pdm09) virus, suggesting a role in host adaptation. To identify NS1 functions regulated by S205 phosphorylation, we generated recombinant PR8 H1N1 NS1 mutants with S205G (nonphosphorylatable) or S205N (H1N1pdm09 signature), as well as H1N1pdm09 viruses harboring the reverse mutation NS1 N205S or N205D (phosphomimetic). Replication of PR8 NS1 mutants was attenuated relative to wild-type (WT) virus replication in a porcine cell line. However, PR8 NS1 S205N showed remarkably higher attenuation than PR8 NS1 S205G in a human cell line, highlighting a potential host-independent advantage of phosphorylatable S205, while an asparagine at this position led to a potential host-specific attenuation. Interestingly, PR8 NS1 S205G did not show polymerase activity-enhancing functions, in contrast to the WT, which can be attributed to diminished interaction with cellular restriction factor DDX21. Analysis of the respective kinase mediating S205 phosphorylation indicated an involvement of casein kinase 2 (CK2). CK2 inhibition significantly reduced the replication of WT viruses and decreased NS1-DDX21 interaction, as observed for NS1 S205G. In summary, NS1 S205 is required for efficient NS1-DDX21 binding, resulting in enhanced viral polymerase activity, which is likely to be regulated by transient phosphorylation.IMPORTANCE Influenza A viruses (IAVs) still pose a major threat to human health worldwide. As a zoonotic virus, IAV can spontaneously overcome species barriers and even reside in new hosts after efficient adaptation. Investigation of the functions of specific adaptational mutations can lead to a deeper understanding of viral replication in specific hosts and can probably help to find new targets for antiviral intervention. In the present study, we analyzed the role of NS1 S205, a phosphorylation site that was reacquired during the circulation of pandemic H1N1pdm09 "swine flu" in the human host. We found that phosphorylation of human H1N1 virus NS1 S205 is mediated by the cellular kinase CK2 and is needed for efficient interaction with human host restriction factor DDX21, mediating NS1-induced enhancement of viral polymerase activity. Therefore, targeting CK2 activity might be an efficient strategy for limiting the replication of IAVs circulating in the human population.


Subject(s)
Influenza A virus/physiology , RNA-Dependent RNA Polymerase/metabolism , Serine/metabolism , Viral Nonstructural Proteins/metabolism , Adaptation, Physiological/genetics , Animals , Casein Kinase II/metabolism , Cell Line , DEAD-box RNA Helicases/metabolism , Host-Pathogen Interactions , Humans , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H1N1 Subtype/metabolism , Influenza A Virus, H1N1 Subtype/physiology , Influenza A virus/genetics , Influenza A virus/metabolism , Mutation , Phosphorylation , Protein Binding , Swine , Viral Nonstructural Proteins/genetics , Virus Replication
10.
J Pathol ; 251(4): 388-399, 2020 08.
Article in English | MEDLINE | ID: mdl-32449525

ABSTRACT

Recently, we established a doxycycline-inducible human tumor necrosis factor alpha (TNFα)-transgenic mouse line, ihTNFtg. Non-induced young and elderly mice showed low but constitutive expression of hTNFα due to promoter leakiness. The persistently present hTNFα stimulated endogenous pro-inflammatory mouse mS100A8/A9 alarmins. Secreted mS100A8/A9 in turn induced the expression and release of mouse mTNFα. The continuous upregulation of pro-inflammatory mTNFα and mS100A8/A9 proteins, due to their mutual expression dependency, gradually led to increased levels in colon tissue and blood. This finally exceeded the threshold levels tolerated by the healthy organism, leading to the onset of intestinal inflammation. Here, recombinant hTNFα functioned as an initial trigger for the development of chronic inflammation. Crossing ihTNFtg mice with S100A9KO mice lacking active S100A8/A9 alarmins or with Rag1KO mice lacking T and B lymphocytes completely abrogated the development of colonic inflammation, despite the still leaky hTNFα promoter. Furthermore, both the intensity of the immune response and the strength of immunosuppressive Treg induction was found to depend on the major histocompatibility complex (MHC) genetic composition. In summary, the onset of intestinal inflammation in elderly mice depends on at least four factors that have to be present simultaneously: TNFα upregulation, S100A8/A9 protein expression, functional T lymphocytes and genetic composition, with the MHC haplotype being of central importance. Only joint action of these factors leads to chronic intestinal inflammation, while absence of any of these determinants abrogates the development of the autoimmune disorder. © 2020 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.


Subject(s)
Calgranulin A/metabolism , Calgranulin B/metabolism , Colitis/genetics , Inflammation/genetics , Major Histocompatibility Complex/genetics , Tumor Necrosis Factor-alpha/metabolism , Alarmins/genetics , Alarmins/metabolism , Animals , Bone Marrow Cells , Calgranulin A/genetics , Calgranulin B/genetics , Colitis/metabolism , Colitis/pathology , Colon/metabolism , Colon/pathology , Haplotypes , Humans , Immunohistochemistry , Inflammation/metabolism , Inflammation/pathology , Mice , Mice, Inbred BALB C , Mice, Transgenic , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/pathology , Tumor Necrosis Factor-alpha/genetics , Up-Regulation
11.
Mol Ther Oncolytics ; 17: 190-204, 2020 Jun 26.
Article in English | MEDLINE | ID: mdl-32346609

ABSTRACT

Recently, we showed that infection of primary lung tumor-bearing mice with oncolytic influenza A viruses (IAVs) led to strong virus-induced tumor cell lysis but also to restoration of immune competence of innate immune cells. Murine B16-F10 melanoma cells are known for their high lung tropism and progressive growth. As these cells are also highly permissive for IAVs, we analyzed their oncolytic and immunomodulatory efficiency against pulmonary B16-F10 lung metastases in vivo. IAV infection abrogated the melanoma-mediated immune suppression in the lung and induced a more than 50% cancer cell lysis. The oncolytic effect reached maximal efficacy 3 days post-infection, but it was not sustained over time. In order to maintain the virus-induced anti-tumor effect, mice with melanoma-derived lung cancers were treated in addition to influenza virus infection with an immune checkpoint inhibitor against programmed death-1 receptor (PD-1). The combined IAV and immune checkpoint inhibition (ICI) therapy resulted in a sustained anti-tumor efficacy, keeping the lung melanoma mass at day 12 of IAV infection still reduced by 50% over the control mice. In conclusion, ICI treatment strongly enhanced the oncolytic effect of influenza virus infection, suggesting that combined treatment is a promising approach against metastatic pulmonary melanoma.

12.
FASEB J ; 33(7): 7799-7809, 2019 07.
Article in English | MEDLINE | ID: mdl-30939249

ABSTRACT

The 4-and-a-half LIM domain protein 2 (FHL2) is a multifunctional adaptor protein that can interact with cell surface receptors, cytosolic adaptor and structural proteins, kinases, and nuclear transcription factors. It is involved in numerous functional activities, including the epithelial-mesenchymal transition, cell proliferation, apoptosis, adhesion, migration, structural stability, and gene expression. Despite this, FHL2-knockout (KO) mice are viable and fertile with no obvious abnormalities, rather suggesting a high capacity for fine-tuning adjustment and functional redundancy of FHL2. Indeed, challenging FHL2-KO cells or mice provided numerous evidences for the great functional significance of FHL2. In recent years, several reviews have been published describing the high capacity of FHL2 to bind diverse proteins as well as the versatile functions of FHL2, emphasizing in particular its role in cardiovascular diseases and carcinogenesis. Here, we view the function of FHL2 from a different perspective. We summarize the published data demonstrating the impact of FHL2 on wound healing and inflammation. FHL2 seems to be involved in numerous steps of these extremely complex and multidirectional but tightly regulated tissue remodeling processes, supporting tissue repair and coordinating inflammation. Deficiency of FHL2 not only slows down ongoing wound healing but also often turns it into a chronic condition.-Wixler, V. The role of FHL2 in wound healing and inflammation.


Subject(s)
Inflammation/physiopathology , LIM-Homeodomain Proteins/physiology , Muscle Proteins/physiology , Transcription Factors/physiology , Wound Healing/physiology , Animals , Chemotaxis, Leukocyte/physiology , Cytokines/physiology , Epithelial-Mesenchymal Transition/physiology , Inflammation/immunology , LIM-Homeodomain Proteins/biosynthesis , LIM-Homeodomain Proteins/deficiency , LIM-Homeodomain Proteins/genetics , Mice , Mice, Knockout , Mice, Transgenic , Muscle Proteins/biosynthesis , Muscle Proteins/deficiency , Muscle Proteins/genetics , Myofibroblasts/physiology , Regeneration/physiology , Signal Transduction/physiology , Transcription Factors/biosynthesis , Transcription Factors/deficiency , Transcription Factors/genetics , Transcription, Genetic/physiology , Up-Regulation
13.
J Cell Sci ; 132(6)2019 03 18.
Article in English | MEDLINE | ID: mdl-30745335

ABSTRACT

The four and a half LIM domains protein 2 (Fhl2) is an adaptor protein capable of mediating protein-protein interactions. Here, we report for the first time phenotypic changes in the brain of Fhl2-deficient mice. We showed that Fhl2 is expressed in neural stem cells, precursors and mature cells of neuronal lineage. Moreover, Fhl2 deficiency leads to delayed neuroblast migration in vivo, premature astroglial differentiation of neural stem cells (NSCs) in vitro, and a gliosis-like accumulation of glial fibrillary acidic protein (GFAP)-positive astrocytes in vivo that substantially increases with age. Collectively, Fhl2-deficiency in the brain interrupts the maintenance and the balanced differentiation of adult NSCs, resulting in preferentially glial differentiation and early exhaustion of the NSC pool required for adult neurogenesis.


Subject(s)
Cell Movement , Glial Fibrillary Acidic Protein/metabolism , LIM-Homeodomain Proteins/metabolism , Muscle Proteins/metabolism , Neural Stem Cells/metabolism , Neurogenesis , Transcription Factors/metabolism , Aging , Animals , Astrocytes/metabolism , Brain/cytology , Brain/metabolism , Cell Differentiation , Cells, Cultured , Gliosis/metabolism , HEK293 Cells , Humans , Mice , Neural Stem Cells/cytology , Neurons/metabolism
14.
Pediatr Rheumatol Online J ; 16(1): 39, 2018 Jun 20.
Article in English | MEDLINE | ID: mdl-29925386

ABSTRACT

BACKGROUND: Inflammatory effector T cells trigger inflammation despite increased numbers of Treg cells in the synovial joint of patients suffering from juvenile idiopathic arthritis (JIA). The cAMP response element (CREM)α is known to play a major role in regulation of T cells in SLE, colitis, and EAE. However, its role in regulation of effector T cells within the inflammatory joint is unknown. METHODS: CREM expression was analyzed in synovial fluid cells from oligoarticular JIA patients by flow cytometry. Peripheral blood mononuclear cells were incubated with synovial fluid and analyzed in the presence and absence of CREM using siRNA experiments for T cell phenotypes. To validate the role of CREM in vivo, ovalbumin-induced T cell dependent arthritis experiments were performed. RESULTS: CREM is highly expressed in synovial fluid T cells and its expression can be induced by treating healthy control PBMCs with synovial fluid. Specifically, CREM is more abundant in CD161+ subsets, than CD161- subsets, of T cells and contributes to cytokine expression by these cells. Finally, development of ovalbumin-induced experimental arthritis is ameliorated in mice with adoptively transferred CREM-/- T cells. CONCLUSION: In conclusion, our study reveals that beyond its role in SLE T cells CREM also drives an inflammatory phenotype of T cells in JIA.


Subject(s)
Arthritis, Juvenile/metabolism , Cyclic AMP Response Element Modulator/metabolism , Synovial Fluid/metabolism , T-Lymphocytes/metabolism , Animals , Cell Culture Techniques , Flow Cytometry/methods , Humans , Joints/pathology , Mice , Phenotype , Real-Time Polymerase Chain Reaction
15.
Oncoimmunology ; 7(5): e1423171, 2018.
Article in English | MEDLINE | ID: mdl-29721377

ABSTRACT

Non-small-cell lung cancer (NSCLC) is the most frequent type of lung cancer and demonstrates high resistance to radiation and chemotherapy. These tumors evade immune system detection by promoting an immunosuppressive tumor microenvironment. Genetic analysis has revealed oncogenic activation of the Ras/Raf/MEK/ERK signaling pathway to be a hallmark of NSCLCs, which promotes influenza A virus (IAV) infection and replication in these cells. Thus, we aimed to unravel the oncolytic properties of IAV infection against NSCLCs in an immunocompetent model in vivo. Using Raf-BxB transgenic mice that spontaneously develop NSCLCs, we demonstrated that infection with low-pathogenic IAV leads to rapid and efficient oncolysis, eliminating 70% of the initial tumor mass. Interestingly, IAV infection of Raf-BxB mice caused a functional reversion of immunosuppressed tumor-associated lung macrophages into a M1-like pro-inflammatory active phenotype that additionally supported virus-induced oncolysis of cancer cells. Altogether, our data demonstrate for the first time in an immunocompetent in vivo model that oncolytic IAV infection is capable of restoring and redirecting immune cell functions within the tumor microenvironment of NSCLCs.

16.
J Clin Invest ; 128(5): 1852-1866, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29611822

ABSTRACT

Autoimmune diseases, such as psoriasis and arthritis, show a patchy distribution of inflammation despite systemic dysregulation of adaptive immunity. Thus, additional tissue-derived signals, such as danger-associated molecular patterns (DAMPs), are indispensable for manifestation of local inflammation. S100A8/S100A9 complexes are the most abundant DAMPs in many autoimmune diseases. However, regulatory mechanisms locally restricting DAMP activities are barely understood. We now unravel for the first time, to our knowledge, a mechanism of autoinhibition in mice and humans restricting S100-DAMP activity to local sites of inflammation. Combining protease degradation, pull-down assays, mass spectrometry, and targeted mutations, we identified specific peptide sequences within the second calcium-binding EF-hands triggering TLR4/MD2-dependent inflammation. These binding sites are free when S100A8/S100A9 heterodimers are released at sites of inflammation. Subsequently, S100A8/S100A9 activities are locally restricted by calcium-induced (S100A8/S100A9)2 tetramer formation hiding the TLR4/MD2-binding site within the tetramer interphase, thus preventing undesirable systemic effects. Loss of this autoinhibitory mechanism in vivo results in TNF-α-driven fatal inflammation, as shown by lack of tetramer formation in crossing S100A9-/- mice with 2 independent TNF-α-transgene mouse strains. Since S100A8/S100A9 is the most abundant DAMP in many inflammatory diseases, specifically blocking the TLR4-binding site of active S100 dimers may represent a promising approach for local suppression of inflammatory diseases, avoiding systemic side effects.


Subject(s)
Alarmins/immunology , Calgranulin A/immunology , Calgranulin B/immunology , Alarmins/chemistry , Alarmins/genetics , Animals , Arthritis/genetics , Arthritis/immunology , Arthritis/pathology , Binding Sites , Calgranulin A/chemistry , Calgranulin A/genetics , Calgranulin B/chemistry , Calgranulin B/genetics , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Mice , Mice, Knockout , Proto-Oncogene Proteins c-mdm2/genetics , Proto-Oncogene Proteins c-mdm2/immunology , Psoriasis/genetics , Psoriasis/immunology , Psoriasis/pathology , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/immunology
17.
Am J Pathol ; 188(5): 1236-1245, 2018 05.
Article in English | MEDLINE | ID: mdl-29458009

ABSTRACT

Four-and-a-half LIM domain protein 2 (FHL2) is a multifunctional adaptor protein with fine-tuning adjustment properties. It acts as a regulator of signaling cascades but also as a cofactor of transcription and controls several anti-inflammatory immune responses. Recently, we described FHL2 as a novel regulator of influenza A virus propagation. We have shown that in vitro FHL2 restricts viral replication by accelerating the interferon regulatory factor 3-dependent transcription of the Ifnb1 gene. In this work, we unraveled an ambiguous role of FHL2 during influenza A virus infection in vivo. Although FHL2 restrained viral replication during the first 24 hours of infection, it significantly delayed viral clearance afterward. Comparison of lung immune status of wild-type and FHL2 knockout mice during influenza virus infection did not acknowledge significant differences in the innate host immune response but revealed an improved migration of dendritic cells from infected lungs into draining lymph nodes as well as increased levels of activated CD8+ T lymphocytes accumulated in the lungs of FHL2 knockout mice.


Subject(s)
Adaptive Immunity/physiology , Inflammation/metabolism , Influenza A virus , LIM-Homeodomain Proteins/metabolism , Lung/metabolism , Muscle Proteins/metabolism , Transcription Factors/metabolism , Animals , Cell Movement/physiology , Dendritic Cells/cytology , Dendritic Cells/metabolism , Inflammation/pathology , Inflammation/virology , LIM-Homeodomain Proteins/genetics , Lung/pathology , Lung/virology , Mice , Mice, Knockout , Muscle Proteins/genetics , Transcription Factors/genetics , Virus Replication/physiology
18.
Arthritis Rheumatol ; 70(3): 371-382, 2018 03.
Article in English | MEDLINE | ID: mdl-29193895

ABSTRACT

OBJECTIVE: Fibroblast-like synoviocytes (FLS) produce key synovial fluid and tissue components to ensure joint integrity under healthy conditions, whereas they become cancer-like and aggressively contribute to joint degeneration in inflammatory arthritis. The aim of this study was to determine whether the SOXC transcription factors SOX4 and SOX11, whose functions are critical in joint development and many cancer types, contribute to FLS activities under normal and inflammatory conditions. METHODS: We inactivated the SOXC genes in FLS from adult mice and studied the effect on joint homeostasis and tumor necrosis factor (TNF)-induced arthritis. We used primary cells and synovial biopsy specimens from arthritis patients to analyze the interactions between inflammatory signals and SOXC proteins. RESULTS: Postnatal inactivation of the SOXC genes had no major effect on joint integrity in otherwise healthy mice. However, it hampered synovial hyperplasia and joint degeneration in transgenic mice expressing human TNF. These effects were explained by the ability of SOX4/11 to amplify the pathogenic impact of TNF on FLS by increasing their survival and migration. SOXC RNA levels were not changed by TNF and other proinflammatory cytokines, but SOXC proteins were strongly stabilized and able to potentiate the TNF-induced up-regulation of genes involved in FLS transformation. Substantiating the relevance of these findings in human disease, SOXC protein levels, but not RNA levels, were significantly higher in inflamed synovium than in noninflamed synovium from arthritis patients. CONCLUSION: SOXC proteins are targets and pivotal mediators of proinflammatory cytokines during FLS transformation in arthritic diseases. Targeting of these proteins could thus improve current strategies to treat arthritic diseases and possibly other inflammatory diseases.


Subject(s)
Arthritis/metabolism , Cytokines/metabolism , SOXC Transcription Factors/metabolism , Synoviocytes/metabolism , Animals , Cell Transformation, Neoplastic/metabolism , Cells, Cultured , Humans , In Situ Hybridization , Mice , Real-Time Polymerase Chain Reaction , Synovial Membrane/metabolism , Tumor Necrosis Factor-alpha/metabolism
19.
Am J Pathol ; 187(11): 2388-2398, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28823868

ABSTRACT

Four and a half LIM domain protein 2 (Fhl2) is an intracellular adaptor molecule with a high protein-protein interaction capacity. It acts as a modulator of several signaling molecules in the cytosol and as a cofactor of transcription in the nucleus. Recent studies suggest the role of Fhl2 in tissue repair and the anti-inflammatory response. Herein, we show that Fhl2-deficient mice develop a more severe psoriatic arthritis disease under induction of the inducible human tumor necrosis factor (hTNF) transgene than wild-type mice. The disease was accompanied by increased infiltration of activated macrophages and T regulatory cells in skin and digit joints as well as by increased expression of matrix metalloproteases and bone-specific proteases. The more severe pathogenesis of psoriatic arthritis in Fhl2 knockout mice coincided with enhanced levels of soluble hTNF cytokine, but surprisingly not with transcription of the hTNF transgene. Studying the shedding of cell membrane-bound hTNF by Adam17, a known Fhl2 interacting protein, revealed an enhanced release of TNF in the absence of Fhl2. In summary, our results show that Fhl2 anticipates the emerging inflammation and specifically the development of psoriatic arthritis by impeding the Adam17-mediated release of TNF.


Subject(s)
ADAM17 Protein/metabolism , Arthritis, Psoriatic/metabolism , LIM Domain Proteins/metabolism , LIM-Homeodomain Proteins/metabolism , Muscle Proteins/metabolism , Transcription Factors/metabolism , Animals , Arthritis, Psoriatic/genetics , Cells, Cultured , Homeodomain Proteins/metabolism , Humans , Inflammation/metabolism , LIM-Homeodomain Proteins/genetics , Mice, Knockout , Muscle Proteins/genetics , Transcription Factors/genetics , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
20.
Cell Microbiol ; 19(7)2017 07.
Article in English | MEDLINE | ID: mdl-28076660

ABSTRACT

Expression of the influenza A virus (IAV) nonstructural protein (NS1) results in the activation of c-Jun N-terminal kinase (JNK). Both NS1 and JNK are involved in apoptosis induction. To investigate their interrelationship, we stably expressed a tamoxifen inducible NS1 oestrogen receptor fusion-protein (NS1ERT) in mammalian cells. Upon tamoxifen stimulation, NS1ERT-expressing cells partially rescued the attenuated replication of NS1-deficient IAVs and also inhibited interferon up-regulation, confirming the functional competence of NS1ERT. Tamoxifen-induced NS1ERT created a cytopathic phenotype and led to the activation of JNK and apoptosis. Induction of NS1F103SERT mutant failed to activate JNK, but induced apoptosis, whereas the induction of NS1M106IERT led to JNK phosphorylation, but not apoptosis, indicating that JNK activation and apoptosis induction are not functionally linked. Further mutational analysis highlighted that apoptosis induction is a function of the C-terminal effector domain of NS1. Finally, IAVs encoding mutant NS1 revealed a modulating effect of NS1 on apoptosis induction in a genuine infection. With respect to apoptogenicity, an NS1 mutant virus that results in a super activation of JNK behaves similarly to the JNK nonactivating virus expressing NS1F103S, thus confirming that NS1-mediated JNK activation and apoptosis induction are also functionally independent from each other in vivo.


Subject(s)
Apoptosis/physiology , JNK Mitogen-Activated Protein Kinases/metabolism , Receptors, Estrogen/genetics , Recombinant Fusion Proteins/genetics , Viral Nonstructural Proteins/genetics , A549 Cells , Animals , Cell Line , Dogs , Enzyme Activation , HEK293 Cells , Humans , Influenza A virus/metabolism , Madin Darby Canine Kidney Cells , Receptors, Estrogen/metabolism , Recombinant Fusion Proteins/metabolism , Tamoxifen/metabolism , Viral Nonstructural Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...