Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 141
Filter
Add more filters

Publication year range
1.
Nat Immunol ; 21(3): 274-286, 2020 03.
Article in English | MEDLINE | ID: mdl-32066947

ABSTRACT

Human immunodeficiency virus 1 (HIV-1) infection is associated with heightened inflammation and excess risk of cardiovascular disease, cancer and other complications. These pathologies persist despite antiretroviral therapy. In two independent cohorts, we found that innate lymphoid cells (ILCs) were depleted in the blood and gut of people with HIV-1, even with effective antiretroviral therapy. ILC depletion was associated with neutrophil infiltration of the gut lamina propria, type 1 interferon activation, increased microbial translocation and natural killer (NK) cell skewing towards an inflammatory state, with chromatin structure and phenotype typical of WNT transcription factor TCF7-dependent memory T cells. Cytokines that are elevated during acute HIV-1 infection reproduced the ILC and NK cell abnormalities ex vivo. These results show that inflammatory cytokines associated with HIV-1 infection irreversibly disrupt ILCs. This results in loss of gut epithelial integrity, microbial translocation and memory NK cells with heightened inflammatory potential, and explains the chronic inflammation in people with HIV-1.


Subject(s)
Cytokines/blood , HIV-1/immunology , HIV-1/pathogenicity , Immunity, Innate , Killer Cells, Natural/immunology , Lymphocytes/immunology , T Cell Transcription Factor 1/immunology , Gene Expression Regulation , HIV Infections/genetics , HIV Infections/immunology , HIV Infections/virology , Homeostasis/immunology , Humans , Immunologic Memory , In Vitro Techniques , Inflammation/genetics , Inflammation/immunology , Inflammation/virology , T Cell Transcription Factor 1/genetics , Wnt Signaling Pathway/immunology
2.
Immunity ; 48(5): 963-978.e3, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29768179

ABSTRACT

Regulated antimicrobial peptide expression in the intestinal epithelium is key to defense against infection and to microbiota homeostasis. Understanding the mechanisms that regulate such expression is necessary for understanding immune homeostasis and inflammatory disease and for developing safe and effective therapies. We used Caenorhabditis elegans in a preclinical approach to discover mechanisms of antimicrobial gene expression control in the intestinal epithelium. We found an unexpected role for the cholinergic nervous system. Infection-induced acetylcholine release from neurons stimulated muscarinic signaling in the epithelium, driving downstream induction of Wnt expression in the same tissue. Wnt induction activated the epithelial canonical Wnt pathway, resulting in the expression of C-type lectin and lysozyme genes that enhanced host defense. Furthermore, the muscarinic and Wnt pathways are linked by conserved transcription factors. These results reveal a tight connection between the nervous system and the intestinal epithelium, with important implications for host defense, immune homeostasis, and cancer.


Subject(s)
Acetylcholine/immunology , Caenorhabditis elegans/immunology , Intestinal Mucosa/immunology , Wnt Signaling Pathway/immunology , Acetylcholine/metabolism , Animals , Antimicrobial Cationic Peptides/genetics , Antimicrobial Cationic Peptides/immunology , Antimicrobial Cationic Peptides/metabolism , Bacteria/immunology , Caenorhabditis elegans/genetics , Caenorhabditis elegans/microbiology , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/immunology , Caenorhabditis elegans Proteins/metabolism , Gene Expression/immunology , Homeostasis/genetics , Homeostasis/immunology , Host-Pathogen Interactions/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Neurons/immunology , Neurons/metabolism , Wnt Signaling Pathway/genetics
3.
J Biol Chem ; 299(11): 105276, 2023 11.
Article in English | MEDLINE | ID: mdl-37739035

ABSTRACT

Imbalanced immune responses are a prominent hallmark of cancer and autoimmunity. Myeloid cells can be overly suppressive, inhibiting protective immune responses or inactive not controlling autoreactive immune cells. Understanding the mechanisms that induce suppressive myeloid cells, such as myeloid-derived suppressor cells (MDSCs) and tolerogenic dendritic cells (TolDCs), can facilitate the development of immune-restoring therapeutic approaches. MDSCs are a major barrier for effective cancer immunotherapy by suppressing antitumor immune responses in cancer patients. TolDCs are administered to patients to promote immune tolerance with the intent to control autoimmune disease. Here, we investigated the development and suppressive/tolerogenic activity of human MDSCs and TolDCs to gain insight into signaling pathways that drive immunosuppression in these different myeloid subsets. Moreover, monocyte-derived MDSCs (M-MDSCs) generated in vitro were compared to M-MDSCs isolated from head-and-neck squamous cell carcinoma patients. PI3K-AKT signaling was identified as being crucial for the induction of human M-MDSCs. PI3K inhibition prevented the downregulation of HLA-DR and the upregulation of reactive oxygen species and MerTK. In addition, we show that the suppressive activity of dexamethasone-induced TolDCs is induced by ß-catenin-dependent Wnt signaling. The identification of PI3K-AKT and Wnt signal transduction pathways as respective inducers of the immunomodulatory capacity of M-MDSCs and TolDCs provides opportunities to overcome suppressive myeloid cells in cancer patients and optimize therapeutic application of TolDCs. Lastly, the observed similarities between generated- and patient-derived M-MDSCs support the use of in vitro-generated M-MDSCs as powerful model to investigate the functionality of human MDSCs.


Subject(s)
Dendritic Cells , Myeloid-Derived Suppressor Cells , Phosphatidylinositol 3-Kinases , Signal Transduction , Wnt Signaling Pathway , Humans , Dendritic Cells/immunology , Immunomodulation/immunology , Immunotherapy , Myeloid-Derived Suppressor Cells/immunology , Neoplasms/immunology , Neoplasms/therapy , Phosphatidylinositol 3-Kinases/immunology , Proto-Oncogene Proteins c-akt/immunology , Signal Transduction/immunology , Wnt Signaling Pathway/immunology , Tumor Cells, Cultured
4.
Gynecol Oncol ; 185: 83-94, 2024 06.
Article in English | MEDLINE | ID: mdl-38377762

ABSTRACT

OBJECTIVE: Advanced-stage high-grade serous ovarian cancer (HGSOC) remains a deadly gynecologic malignancy with high rates of disease recurrence and limited, effective therapeutic options for patients. There is a significant need to better stratify HGSOC patients into platinum refractory (PRF) vs. sensitive (PS) cohorts at baseline to improve therapeutic responses and survival outcomes for PRF HGSOC. METHODS: We performed NanoString for GeoMx Digital Spatial Profile (G-DSP) multiplex protein analysis on PRF and PS tissue microarrays (TMAs) to study the bidirectional communication of cancer cells with immune cells in the tumor microenvironment (TME) of HGSOC. We demonstrate robust stratification of PRF and PS tumors at baseline using multiplex spatial proteomic biomarkers with implications for tailoring subsequent therapy. RESULTS: PS patients had elevated apoptotic and anti-tumor immune profiles, while PRF patients had dual AKT1 and WNT signaling with immunosuppressive profiles. We found that dual activity of AKT1 and WNT signaling supported the exclusion of immune cells, specifically tumor infiltrating lymphocytes (TILs), from the TME in PRF tumors, and this was not observed in PS tumors. The exclusion of immune cells from the TME of PRF tumors corresponded to abnormal endothelial cell structure in tumors with dual AKT1 and WNT signaling activity. CONCLUSIONS: We believe our findings provide improved understanding of tumor-immune crosstalk in HGSOC TME highlighting the importance of the relationship between AKT and WNT pathways, immune cell function, and platinum response in HGSOC.


Subject(s)
Drug Resistance, Neoplasm , Ovarian Neoplasms , Proteomics , Proto-Oncogene Proteins c-akt , Tumor Microenvironment , Humans , Female , Tumor Microenvironment/immunology , Ovarian Neoplasms/immunology , Ovarian Neoplasms/pathology , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/drug therapy , Proto-Oncogene Proteins c-akt/metabolism , Proteomics/methods , Drug Resistance, Neoplasm/immunology , Middle Aged , Cystadenocarcinoma, Serous/immunology , Cystadenocarcinoma, Serous/pathology , Cystadenocarcinoma, Serous/drug therapy , Cystadenocarcinoma, Serous/metabolism , Wnt Signaling Pathway/immunology , Aged , Lymphocytes, Tumor-Infiltrating/immunology
5.
Hepatology ; 74(2): 987-1002, 2021 08.
Article in English | MEDLINE | ID: mdl-33619773

ABSTRACT

BACKGROUND AND AIMS: Acute liver damage causes hepatocyte stress and death, but in chronic liver disease impaired hepatocyte regeneration and immune cell infiltration prevents recovery. While the roles of both impaired liver regeneration and immune infiltration have been studied extensively in chronic liver diseases, the differential contribution of these factors is difficult to assess. APPROACH AND RESULTS: We combined single-cell RNA-sequencing (RNA-seq) data from healthy livers and peripheral immune cells to measure cell proportions in chronic liver diseases. Using bulk RNA-seq data from patients with early alcohol-associated hepatitis, severe AH (sAH), HCV, HCV with cirrhosis, and NAFLD, we performed gene deconvolution to predict the contribution of different cell types in each disease. Patients with sAH had the greatest change in cell composition, with increases in both periportal hepatocytes and cholangiocyte populations. Interestingly, while central vein hepatocytes were decreased, central vein endothelial cells were expanded. Endothelial cells are thought to regulate liver regeneration through WNT signaling. WNT2, important in central vein hepatocyte development, was down in sAH, while multiple other WNTs and WNT receptors were up-regulated. Immunohistochemistry revealed up-regulation of FZD6, a noncanonical WNT receptor, in hepatocytes in sAH. Immune cell populations also differed in disease. In sAH, a specific group of inflammatory macrophages was increased and distinct from the macrophage population in patients with HCV. Network and correlation analyses revealed that changes in the cell types in the liver were highly correlated with clinical liver function tests. CONCLUSIONS: These results identify distinct changes in the liver cell populations in chronic liver disease and illustrate the power of using single-cell RNA-seq data from a limited number of samples in understanding multiple different diseases.


Subject(s)
Gene Expression Regulation/immunology , Hepatitis, Alcoholic/immunology , Liver Regeneration/genetics , Liver/pathology , Case-Control Studies , Cluster Analysis , Endothelial Cells/immunology , Endothelial Cells/pathology , Healthy Volunteers , Hepatitis, Alcoholic/genetics , Hepatitis, Alcoholic/pathology , Hepatocytes/immunology , Hepatocytes/pathology , Humans , Liver/immunology , Liver Regeneration/immunology , Macrophages/immunology , Macrophages/metabolism , Monocytes/immunology , Monocytes/metabolism , RNA-Seq , Single-Cell Analysis , Wnt Signaling Pathway/genetics , Wnt Signaling Pathway/immunology
6.
J Immunol ; 205(8): 2265-2275, 2020 10 15.
Article in English | MEDLINE | ID: mdl-32917787

ABSTRACT

Loss of immune tolerance to gut microflora is inextricably linked to chronic intestinal inflammation and colitis-associated colorectal cancer (CAC). The LRP5/6 signaling cascade in APCs contributes to immune homeostasis in the gut, but whether this pathway in APCs protects against CAC is not known. In the current study, using a mouse model of CAC, we show that the LRP5/6-ß-catenin-IL-10 signaling axis in intestinal CD11c+ APCs protects mice from CAC by regulating the expression of tumor-promoting inflammatory factors in response to commensal flora. Genetic deletion of LRP5/6 in CD11c+ APCs in mice (LRP5/6ΔCD11c) resulted in enhanced susceptibility to CAC. This is due to a microbiota-dependent increased expression of proinflammatory factors and decreased expression of the immunosuppressive cytokine IL-10. This condition could be improved in LRP5/6ΔCD11c mice by depleting the gut flora, indicating the importance of LRP5/6 in mediating immune tolerance to the gut flora. Moreover, mechanistic studies show that LRP5/6 suppresses the expression of tumor-promoting inflammatory factors in CD11c+ APCs via the ß-catenin-IL-10 axis. Accordingly, conditional activation of ß-catenin specifically in CD11c+ APCs or in vivo administration of IL-10 protected LRP5/6ΔCD11c mice from CAC by suppressing the expression of inflammatory factors. In summary, in this study, we identify a key role for the LRP5/6-ß-catenin-IL-10 signaling pathway in intestinal APCs in resolving chronic intestinal inflammation and protecting against CAC in response to the commensal flora.


Subject(s)
Antigen-Presenting Cells/immunology , Colitis/immunology , Colonic Neoplasms/immunology , Gastrointestinal Microbiome/immunology , Interleukin-10/immunology , Wnt Signaling Pathway/immunology , beta Catenin/immunology , Animals , Antigen-Presenting Cells/pathology , Colitis/complications , Colitis/genetics , Colitis/pathology , Colonic Neoplasms/etiology , Colonic Neoplasms/genetics , Colonic Neoplasms/prevention & control , Gastrointestinal Microbiome/genetics , Interleukin-10/genetics , Mice , Mice, Transgenic , Neoplasm Proteins/genetics , Wnt Signaling Pathway/genetics , beta Catenin/genetics
7.
J Immunol ; 205(8): 2046-2055, 2020 10 15.
Article in English | MEDLINE | ID: mdl-32887752

ABSTRACT

CD8+ T cells do not rely solely on cytotoxic functions for significant HIV control. Moreover, the noncytotoxic CD8+ T cell antiviral response is a primary mediator of natural HIV control such as that seen in HIV elite controllers and long-term nonprogressors that does not require combined antiretroviral therapy. In this study, we investigated the biological factors contributing to the noncytotoxic control of HIV replication mediated by primary human CD8+ T cells. We report that canonical Wnt signaling inhibits HIV transcription in an MHC-independent, noncytotoxic manner and that mediators of this pathway correlate with HIV controller clinical status. We show that CD8+ T cells express all 19 Wnts and CD8+ T cell-conditioned medium (CM) induced canonical Wnt signaling in infected recipient cells while simultaneously inhibiting HIV transcription. Antagonizing canonical Wnt activity in CD8+ T cell CM resulted in increased HIV transcription in infected cells. Further, Wnt2b expression was upregulated in HIV controllers versus viremic patients, and in vitro depletion of Wnt2b and/or Wnt9b from CD8+ CM reversed HIV inhibitory activity. Finally, plasma concentration of Dkk-1, an antagonist of canonical Wnt signaling, was higher in viremic patients with lower CD4 counts. This study demonstrates that canonical Wnt signaling inhibits HIV and significantly correlates with HIV controller status.


Subject(s)
CD8-Positive T-Lymphocytes , Gene Expression Regulation/immunology , Glycoproteins , HIV Infections , HIV-1 , Immunity, Cellular , Wnt Proteins , Wnt Signaling Pathway/immunology , Adult , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/pathology , Female , Glycoproteins/blood , Glycoproteins/immunology , HIV Infections/blood , HIV Infections/immunology , HIV Infections/pathology , HIV-1/immunology , HIV-1/metabolism , Humans , Intercellular Signaling Peptides and Proteins/blood , Intercellular Signaling Peptides and Proteins/immunology , Male , Wnt Proteins/blood , Wnt Proteins/immunology
8.
Cancer Metastasis Rev ; 39(3): 933-952, 2020 09.
Article in English | MEDLINE | ID: mdl-32435939

ABSTRACT

Wnt5a is the prototypical activator of the non-canonical Wnt pathways, and its overexpression has been implicated in the progression of several tumor types by promoting cell motility, invasion, EMT, and metastasis. Recent evidences have revealed a novel role of Wnt5a in the phosphorylation of the NF-κB subunit p65 and the activation of the NF-κB pathway in cancer cells. In this article, we review the molecular mechanisms and mediators defining a Wnt5a/NF-κB signaling pathway and propose that the aberrant expression of Wnt5a in some tumors drives a Wnt5a/NF-κB/IL-6/STAT3 positive feedback loop that amplifies the effects of Wnt5a. The evidences discussed here suggest that Wnt5a has a double effect on the tumor microenvironment. First, it activates an autocrine ROR1/Akt/p65 pathway that promotes inflammation and chemotaxis of immune cells. Then, Wnt5a activates a TLR/MyD88/p50 pathway exclusively in myelomonocytic cells promoting the synthesis of the anti-inflammatory cytokine IL-10 and a tolerogenic phenotype. As a result of these mechanisms, Wnt5a plays a negative role on immune cell function that contributes to an immunosuppressive tumor microenvironment and would contribute to resistance to immunotherapy. Finally, we summarized the development of different strategies targeting either Wnt5a or the Wnt5a receptor ROR1 that can be helpful for cancer therapy by contributing to generate a more immunostimulatory tumor microenvironment.


Subject(s)
Neoplasms/immunology , Wnt Signaling Pathway/immunology , Wnt-5a Protein/immunology , Animals , Humans , Immune Tolerance , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , NF-kappa B/immunology , NF-kappa B/metabolism , Neoplasm Metastasis , Neoplasms/metabolism , Neoplasms/pathology , Tumor Microenvironment/immunology , Wnt-5a Protein/metabolism
9.
Cell Immunol ; 361: 104273, 2021 03.
Article in English | MEDLINE | ID: mdl-33422699

ABSTRACT

Much attention has been paid to a newly discovered subset of memory T (TM) cells-stem cell-like memory T (TSCM) cells for their high self-renewal ability, multi-differentiation potential and long-term effector function in adoptive therapy against tumors. Despite their application in cancer therapy, an excess of TSCM cells also contributes to the persistence of autoimmune diseases for their immune memory and HIV infection as a long-lived HIV reservoir. Signaling pathways Wnt, AMPK/mTOR and NF-κB are key determinants for TM cell generation, maintenance and proinflammatory effect. In this review, we focus on the phenotypic and functional characteristics of TSCM cells and discuss their role in autoimmune diseases and HIV-1 chronic infection. Also, we explore the potential mechanism and signaling pathways involved in immune memory and look into the future therapy strategies of targeting long-lived TM cells to suppress pathogenic immune memory.


Subject(s)
Immunologic Memory/immunology , Stem Cells/immunology , T-Lymphocytes/immunology , Adenylate Kinase/immunology , Autoimmune Diseases/immunology , CD4-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , HIV Infections/immunology , Humans , Signal Transduction/immunology , TOR Serine-Threonine Kinases/immunology , Wnt Signaling Pathway/immunology
10.
BMC Cancer ; 21(1): 625, 2021 May 27.
Article in English | MEDLINE | ID: mdl-34044804

ABSTRACT

BACKGROUND: Newcastle disease virus (NDV) is an oncolytic virus with excellent selectivity against cancer cells, both in vitro and in vivo. Unfortunately, prolonged in vitro NDV infection results in the development of persistent infection in the cancer cells which are then able to resist NDV-mediated oncolysis. However, the mechanism of persistency of infection remains poorly understood. METHODS: In this study, we established persistently NDV-infected EJ28 bladder cancer cells, designated as EJ28P. Global transcriptomic analysis was subsequently carried out by microarray analysis. Differentially expressed genes (DEGs) between EJ28 and EJ28P cells identified by the edgeR program were further analysed by Gene Set Enrichment Analysis (GSEA) and Ingenuity Pathway Analysis (IPA) analyses. In addition, the microarray data were validated by RT-qPCR. RESULTS: Persistently NDV-infected EJ28 bladder cancer cells were successfully established and confirmed by flow cytometry. Microarray analysis identified a total of 368 genes as differentially expressed in EJ28P cells when compared to the non-infected EJ28 cells. GSEA revealed that the Wnt/ß-catenin and KRAS signalling pathways were upregulated while the TGF-ß signalling pathway was downregulated. Findings from this study suggest that the upregulation of genes that are associated with cell growth, pro-survival, and anti-apoptosis may explain the survivability of EJ28P cells and the development of persistent infection of NDV. CONCLUSIONS: This study provides insights into the transcriptomic changes that occur and the specific signalling pathways that are potentially involved in the development and maintenance of NDV persistency of infection in bladder cancer cells. These findings warrant further investigation and is crucial towards the development of effective NDV oncolytic therapy against cancer.


Subject(s)
Newcastle disease virus/immunology , Oncolytic Virotherapy/methods , Oncolytic Viruses/immunology , Urinary Bladder Neoplasms/therapy , Cell Line, Tumor , Cell Survival/genetics , Cell Survival/immunology , Down-Regulation/immunology , Gene Expression Regulation, Neoplastic/immunology , Humans , Proto-Oncogene Proteins p21(ras)/metabolism , Transforming Growth Factor beta/metabolism , Up-Regulation/immunology , Urinary Bladder/immunology , Urinary Bladder/pathology , Urinary Bladder Neoplasms/immunology , Wnt Signaling Pathway/genetics , Wnt Signaling Pathway/immunology , beta Catenin/metabolism
11.
Gynecol Oncol ; 160(1): 285-294, 2021 01.
Article in English | MEDLINE | ID: mdl-33168307

ABSTRACT

OBJECTIVE: Immune checkpoint blockade (ICB) therapy shows limited efficacy in ovarian cancers due to the "cold" immune phenotype surrounding these tumors. Previous studies have shown that in ovarian cancer Wnt/ß-catenin pathway activation contributes to this immune phenotype. Here, we evaluated the anti-tumor and immune-enhancing properties of the Wnt inhibitor, CGX-1321, used alone or in combination with either DKN-01 or anti-PD-1 therapy, in pre-clinical ovarian cancer models. METHODS: The parental ID8 murine ovarian cancer model harboring a knock-out of p53 (ID8p53-/-) and MISIIR-Tag spontaneous ovarian cancer models were used to test the effects of CGX-1321 alone or in combination therapies on tumor burden and immune cell landscape in the tumor microenvironment (TME). Flow cytometry and NanoString analyses were used to characterize the changes in tumor-intrinsic signaling and immune-related profiles in the TME of ovarian cancer in response to treatments. RESULTS: CGX-1321 significantly reduced tumor burden and constrained tumor progression in the ID8p53-/- and MISIIR-Tag models. Furthermore, CGX-1321 increased infiltrating CD8+ T cells in the TME. Combining CGX-1321 with either DKN-01 or anti-PD-1 therapy also decreased tumor burden and increased CD8+ T cell infiltration in the omentum TME but did not do so to a greater extent that CGX-1321 monotherapy. CONCLUSIONS: CGX-1321 significantly reduced tumor burden and enhanced CD8+ T cell levels in ovarian cancer, nevertheless the addition of DKN-01 or anti-PD-1 therapies did not enhance these effects of CGX-1321. Further investigation is needed to determine if CGX-1321 + DKN-01 combination treatment sensitizes pre-clinical ovarian cancer to ICB therapy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Enzyme Inhibitors/pharmacology , Immune Checkpoint Inhibitors/pharmacology , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/immunology , Wnt Signaling Pathway/drug effects , Wnt Signaling Pathway/immunology , Acyltransferases/antagonists & inhibitors , Acyltransferases/metabolism , Animals , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Drug Synergism , Enzyme Inhibitors/administration & dosage , Female , Immune Checkpoint Inhibitors/administration & dosage , Intercellular Signaling Peptides and Proteins/metabolism , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/immunology , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Ovarian Neoplasms/metabolism , Tumor Microenvironment , Wnt Proteins/antagonists & inhibitors , Wnt Proteins/metabolism , beta Catenin/metabolism
12.
J Immunol ; 202(8): 2296-2306, 2019 04 15.
Article in English | MEDLINE | ID: mdl-30814306

ABSTRACT

NK cells are innate-like lymphocytes that eliminate virally infected and cancerous cells, but the mechanisms that control NK cell development and cytotoxicity are incompletely understood. We identified roles for sclerostin domain-containing-1 (Sostdc1) in NK cell development and function. Sostdc1-knockout (Sostdc1 -/-) mice display a progressive accumulation of transitional NK cells (tNKs) (CD27+CD11b+) with age, indicating a partial developmental block. The NK cell Ly49 repertoire in Sostdc1 -/- mice is also changed. Lower frequencies of Sostdc1 -/- splenic tNKs express inhibitory Ly49G2 receptors, but higher frequencies express activating Ly49H and Ly49D receptors. However, the frequencies of Ly49I+, G2+, H+, and D+ populations were universally decreased at the most mature (CD27-CD11b+) stage. We hypothesized that the Ly49 repertoire in Sostdc1 -/- mice would correlate with NK killing ability and observed that Sostdc1-/- NK cells are hyporesponsive against MHC class I-deficient cell targets in vitro and in vivo, despite higher CD107a surface levels and similar IFN-γ expression to controls. Consistent with Sostdc1's known role in Wnt signaling regulation, Tcf7 and Lef1 levels were higher in Sostdc1 -/- NK cells. Expression of the NK development gene Id2 was decreased in Sostdc1-/- immature NK and tNK cells, but Eomes and Tbx21 expression was unaffected. Reciprocal bone marrow transplant experiments showed that Sostdc1 regulates NK cell maturation and expression of Ly49 receptors in a cell-extrinsic fashion from both nonhematopoietic and hematopoietic sources. Taken together, these data support a role for Sostdc1 in the regulation of NK cell maturation and cytotoxicity, and identify potential NK cell niches.


Subject(s)
Bone Morphogenetic Proteins/immunology , Immunity, Cellular , Killer Cells, Natural/immunology , Wnt Signaling Pathway/immunology , Adaptor Proteins, Signal Transducing , Animals , Bone Morphogenetic Proteins/genetics , Hepatocyte Nuclear Factor 1-alpha/genetics , Hepatocyte Nuclear Factor 1-alpha/immunology , Inhibitor of Differentiation Protein 2/genetics , Inhibitor of Differentiation Protein 2/immunology , Killer Cells, Natural/cytology , Lymphoid Enhancer-Binding Factor 1/genetics , Lymphoid Enhancer-Binding Factor 1/immunology , Mice , Mice, Knockout , T-Box Domain Proteins/genetics , T-Box Domain Proteins/immunology , Wnt Signaling Pathway/genetics
13.
J Immunol ; 203(10): 2630-2643, 2019 11 15.
Article in English | MEDLINE | ID: mdl-31586036

ABSTRACT

Crlz-1 was expressed along with Wnt3a in the rapidly proliferating centroblasts within the dark zone of germinal center (GC) during humoral immune responses. Significantly, Crlz-1 relayed a Wnt/ß-catenin signal to the expression of Bcl-6, the master regulator of centroblasts, by mobilizing the cytoplasmic CBFß into the nucleus to allow Runx/CBFß heterodimerization and its subsequent binding to the Bcl-6 promoter. The knockdown of Crlz-1 or ß-catenin, as well as inhibition of Wnt signaling in the centroblasts, led to the decreased expression of Bcl-6 and, thereby, the altered expression of its various target genes, resulting in their diminished proliferation. Consistently, the administration of Wnt inhibitors into the immunized mice impaired or abolished GC reaction, with concomitant decreases of Crlz-1 and Bcl-6 expression and, thus, centroblastic proliferation. Our observation that Wnt/ß-catenin signaling via Crlz-1 regulates GC reaction would suggest developmental strategies for vaccine adjuvants and cancer therapeutics because both immune efficacy and accidental lymphoma depend on GC reaction. Our studies of Crlz-1 were performed using human cell lines, mice, and their primary cells.


Subject(s)
B-Lymphocytes/immunology , DNA-Binding Proteins/metabolism , Germinal Center/immunology , Immunity, Humoral , Nerve Tissue Proteins/metabolism , Proto-Oncogene Proteins c-bcl-6/metabolism , Trans-Activators/metabolism , Transcription Factors/metabolism , Wnt Signaling Pathway/immunology , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Mice , Mice, Inbred BALB C , Transfection , Wnt3A Protein/metabolism , beta Catenin/metabolism
14.
Int J Med Sci ; 18(13): 2814-2827, 2021.
Article in English | MEDLINE | ID: mdl-34220309

ABSTRACT

Lower limbs venous insufficiency refers to a wide variety of venous disorders grouped by the term of chronic venous disease (CVD). Hemodynamic and hormonal changes related to pregnancy period, may promote the development of CVD affecting approximately 1 in 3 women. It has been shown that the presence of this condition is associated with damage and placental suffering. Thus, taking IGF-1/PAPP-A/STC-2, inflammatory cytokines production, PI3K/Akt and Wnt/ ß-catenin pathways as a part of the alterations that occurs in the placenta due to CVD, the aim of this study will be to examine the main components of these pathways. Genic and protein expression of PAPP-A, STC-2, IGF-1, IRS-4 Wnt-1, ß-catenin, c-myc, Cyclin D1, IL-4/IL-6 and PI3K/Akt/mTOR pathway will be analysed through RT-qPCR and immunohistochemical techniques in women with CVD (n=62) and pregnant women without this condition (HC) (n=52). PAPP-A, IGF-1, IL-4, IL-6, IRS-4, PI3K, Akt, mTOR, Wnt-1, ß-catenin, c-myc and Cyclin D1 expression were found to be increased in women with CVD, whereas STC-2 were decreased in this group, compared to non-affected women. Our study has demonstrated that IGF-1/PAPP-A/STC-2 axis, PI3K/Akt and Wnt/ß-catenin pathways, along with c-myc, Cyclin D1 and inflammatory cytokines are altered in placenta women with CVD. These results extent the knowledge that CVD is associated to a placenta damage with abnormal tissue environment and cellular regulation.


Subject(s)
Placenta/pathology , Pregnancy Complications, Cardiovascular/immunology , Venous Insufficiency/immunology , Wnt Signaling Pathway/immunology , Adult , Chronic Disease , Female , Glycoproteins/metabolism , Humans , Insulin-Like Growth Factor I/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Placenta/cytology , Pregnancy , Pregnancy Complications, Cardiovascular/pathology , Pregnancy-Associated Plasma Protein-A/metabolism , Prospective Studies , Venous Insufficiency/pathology , Young Adult , beta Catenin/metabolism
15.
Int J Mol Sci ; 22(22)2021 Nov 15.
Article in English | MEDLINE | ID: mdl-34830209

ABSTRACT

The tumor microenvironment (TME) includes immune (T, B, NK, dendritic), stromal, mesenchymal, endothelial, adipocytic cells, extracellular matrix, and cytokines/chemokines/soluble factors regulating various intracellular signaling pathways (ISP) in tumor cells. TME influences the survival/progression of prostate cancer (PC), enabling tumor cell immune-evasion also through the activation of the PD-1/PD-L1 axis. We have performed a systematic literature review according to the PRISMA guidelines, to investigate how the PD-1/PD-L1 pathway is influenced by TME and ISPs. Tumor immune-escape mechanisms include suppression/exhaustion of tumor infiltrating cytotoxic T lymphocytes, inhibition of tumor suppressive NK cells, increase in immune-suppressive immune cells (regulatory T, M2 macrophagic, myeloid-derived suppressor, dendritic, stromal, and adipocytic cells). IFN-γ (the most investigated factor), TGF-ß, TNF-α, IL-6, IL-17, IL-15, IL-27, complement factor C5a, and other soluble molecules secreted by TME components (and sometimes increased in patients' serum), as well as and hypoxia, influenced the regulation of PD-L1. Experimental studies using human and mouse PC cell lines (derived from either androgen-sensitive or androgen-resistant tumors) revealed that the intracellular ERK/MEK, Akt-mTOR, NF-kB, WNT and JAK/STAT pathways were involved in PD-L1 upregulation in PC. Blocking the PD-1/PD-L1 signaling by using immunotherapy drugs can prevent tumor immune-escape, increasing the anti-tumor activity of immune cells.


Subject(s)
B7-H1 Antigen/metabolism , Prostatic Neoplasms/immunology , Prostatic Neoplasms/metabolism , Tumor Microenvironment/immunology , Wnt Signaling Pathway/immunology , Animals , B7-H1 Antigen/antagonists & inhibitors , Cell Line, Tumor , Cytokines/metabolism , Humans , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Immunotherapy/methods , Killer Cells, Natural/immunology , Male , Mice , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/metabolism , Prostatic Neoplasms/therapy , T-Lymphocytes, Cytotoxic/immunology , Tumor Escape , Tumor Microenvironment/drug effects , Wnt Signaling Pathway/drug effects
16.
Cytokine ; 136: 155287, 2020 12.
Article in English | MEDLINE | ID: mdl-32950027

ABSTRACT

Haemophilus parasuis induces severe acute systemic infection in pigs, characterized by fibrinous polyserositis, polyarthritis and meningitis. Our previous study demonstrated that H. parasuis induced the activation of p38 mitogen-activated protein kinase (MAPK) pathway, increasing the expression of proinflammatory genes and mediating H. parasuis-induced inflammation. Moreover, Wnt/ß-catenin signaling activation induced by H. parasuis disrupts the adherens junction between epithelial cells and initiates the epithelial-mesenchymal transition (EMT). In the present study, p38 MAPK was found to be involved in the accumulation of nuclear location of ß-catenin during H. parasuis infection in PK-15 and NPTr cells, via modulating the expression of dickkofp-1 (DKK-1), a negative regulator of Wnt/ß-catenin signaling. We generated DKK-1 knockout cell lines by CRISPR/Cas9-mediated genome editing in PK-15 and NPTr cells, and found that knockout of DKK-1 led to the dysfunction of p38 MAPK in regulating Wnt/ß-catenin signaling activity in H. parasuis-infected cells. Furthermore, p38 MAPK activity was independent of the activation of Wnt/ß-catenin signaling during H. parasuis infection. This is the first study to explore the crosstalk between p38 MAPK and Wnt/ß-catenin signaling during H. parasuis infection. It provides a more comprehensive view of intracellular signaling pathways during pathogenic bacteria-induced acute inflammation.


Subject(s)
Haemophilus Infections , Haemophilus parasuis/immunology , Intercellular Signaling Peptides and Proteins/immunology , Swine Diseases , Swine/immunology , Wnt Signaling Pathway/immunology , p38 Mitogen-Activated Protein Kinases/immunology , Animals , Cell Line , Haemophilus Infections/immunology , Haemophilus Infections/microbiology , Haemophilus Infections/veterinary , Swine/microbiology , Swine Diseases/immunology , Swine Diseases/microbiology
17.
Cytokine ; 136: 155292, 2020 12.
Article in English | MEDLINE | ID: mdl-32950809

ABSTRACT

Osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs) plays a crucial role in osteoporosis. Irisin, an exercise-induced muscle-dependent myokine, has been reported to stimulate the development of brown adipose tissue and regulate energy expenditure. The present study aimed to investigate the effects of irisin on autophagy in BMSCs. Furthermore, the osteogenic differentiation ability was evaluated, as well as the activation of autophagy. It was found that 40 µM irisin for 48 h was an appropriate concentration and time period, with regards to cell viability, which was measured with a Cell Counting Kit-8. Moreover, the increasing expression levels of microtubule-associated protein light chain 3 (Lc3)-I/II and autophagy related 5 (Atg5) by irisin demonstrated the upregulation of autophagy. Mechanistically, bafilomycin A1 and Atg5 small interfering RNA were used to evaluate the possible mechanism of autophagy activated by irisin, and it was identified that irisin may upregulate autophagy by increasing the Atg12-Atg5-Atg16L complex. In addition, with the increasing level of autophagy, osteogenesis and the Wnt/ß-catenin signal pathway were also enhanced. However, inhibition of autophagy by bafilomycin A1 negatively regulated osteogenic differentiation. Collectively, the present results suggested that irisin may stimulate autophagy in BMSCs and that osteogenic differentiation may be enhanced by stimulating autophagy.


Subject(s)
Autophagy/immunology , Bone Marrow Cells/immunology , Cell Differentiation/immunology , Fibronectins/immunology , Mesenchymal Stem Cells/immunology , Osteogenesis/immunology , Wnt Signaling Pathway/immunology , Animals , Mice
18.
J Immunol ; 201(8): 2193-2200, 2018 10 15.
Article in English | MEDLINE | ID: mdl-30301837

ABSTRACT

T cell factor, the effector transcription factor of the WNT signaling pathway, was so named because of the primary observation that it is indispensable for T cell development in the thymus. Since this discovery, the role of this signaling pathway has been extensively studied in T cell development, hematopoiesis, and stem cells; however, its functional role in mature T cells has remained relatively underinvestigated. Over the last few years, various studies have demonstrated that T cell factor can directly influence T cell function and the differentiation of Th1, Th2, Th17, regulatory T cell, follicular helper CD4+ T cell subsets, and CD8+ memory T cells. In this paper, we discuss the molecular mechanisms underlying these observations and place them in the general context of immune responses. Furthermore, we explore the implications and limitations of these findings for WNT manipulation as a therapeutic approach for treating immune-related diseases.


Subject(s)
T-Lymphocyte Subsets/immunology , T-Lymphocytes/immunology , Wnt Signaling Pathway/immunology , Animals , Cell Differentiation , Cytokines/metabolism , Humans , Immunologic Memory , Lymphocyte Activation , TCF Transcription Factors/metabolism
19.
J Immunol ; 201(7): 1855-1864, 2018 10 01.
Article in English | MEDLINE | ID: mdl-30135183

ABSTRACT

Asthma is a syndrome with multifactorial causes, resulting in a variety of different phenotypes. Current treatment options are not curative and are sometimes ineffective in certain disease phenotypes. Therefore, novel therapeutic approaches are required. Recent findings have shown that activation of the canonical Wnt signaling pathway suppresses the development of allergic airway disease. In contrast, the effect of the noncanonical Wnt signaling pathway activation on allergic airway disease is not well described. The aim of this study was to validate the therapeutic effectiveness of Wnt-1-driven canonical Wnt signaling compared with Wnt-5a-driven noncanonical signaling in murine models. In vitro, both ligands were capable of attenuating allergen-specific T cell activation in a dendritic cell-dependent manner. In addition, the therapeutic effects of Wnt ligands were assessed in two different models of allergic airway disease. Application of Wnt-1 resulted in suppression of airway inflammation as well as airway hyperresponsiveness and mucus production. In contrast, administration of Wnt-5a was less effective in reducing airway inflammation or goblet cell metaplasia. These results suggest an immune modulating function for canonical as well as noncanonical Wnt signaling, but canonical Wnt pathway activation appears to be more effective in suppressing allergic airway disease than noncanonical Wnt activation.


Subject(s)
Asthma/immunology , Dendritic Cells/immunology , Respiratory Hypersensitivity/immunology , T-Lymphocytes/immunology , Wnt Signaling Pathway/immunology , Wnt-5a Protein/metabolism , Wnt1 Protein/metabolism , Allergens/immunology , Animals , Cells, Cultured , Disease Models, Animal , Female , Humans , Immunomodulation , Lymphocyte Activation , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Ovalbumin/immunology
20.
J Immunol ; 200(9): 3259-3268, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29602775

ABSTRACT

Aberrant Wnt/ß-catenin signaling occurs in several inflammatory diseases, including inflammatory bowel disease and inflammatory bowel disease-associated colon carcinogenesis. However, its role in shaping mucosal immune responses to commensals in the gut remains unknown. In this study, we investigated the importance of canonical Wnt signaling in CD11c+ APCs in controlling intestinal inflammation. Using a mouse model of ulcerative colitis, we demonstrated that canonical Wnt signaling in intestinal CD11c+ APCs controls intestinal inflammation by imparting an anti-inflammatory phenotype. Genetic deletion of Wnt coreceptors, low-density lipoprotein receptor-related proteins 5 and 6 (LRP5/6) in CD11c+ APCs in LRP5/6ΔCD11c mice, resulted in enhanced intestinal inflammation with increased histopathological severity of colonic tissue. This was due to microbiota-dependent increased production of proinflammatory cytokines and decreased expression of immune-regulatory factors such as IL-10, retinoic acid, and IDO. Mechanistically, loss of LRP5/6-mediated signaling in CD11c+ APCs resulted in altered microflora and T cell homeostasis. Furthermore, our study demonstrates that conditional activation of ß-catenin in CD11c+ APCs in LRP5/6ΔCD11c mice resulted in reduced intestinal inflammation with decreased histopathological severity of colonic tissue. These results reveal a mechanism by which intestinal APCs control intestinal inflammation and immune homeostasis via the canonical Wnt-signaling pathway.


Subject(s)
Antigen-Presenting Cells/immunology , Gastrointestinal Microbiome/immunology , Immunity, Mucosal/immunology , Intestinal Mucosa/immunology , Wnt Signaling Pathway/immunology , Animals , Colitis, Ulcerative/immunology , Colitis, Ulcerative/microbiology , Colon/immunology , Colon/microbiology , Homeostasis/immunology , Inflammation/immunology , Intestinal Mucosa/microbiology , Mice , Mice, Inbred C57BL , Mice, Transgenic
SELECTION OF CITATIONS
SEARCH DETAIL