Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 119
Filter
2.
J Immunother Cancer ; 10(7)2022 07.
Article in English | MEDLINE | ID: mdl-35793868

ABSTRACT

BACKGROUND: Understanding the mechanisms of non-T cell inflamed tumor microenvironment (TME) and their modulation are important to improve cancer immunotherapies such as immune checkpoint inhibitors. The involvement of various immunometabolisms has recently been indicated in the formation of immunosuppressive TME. In this study, we investigated the immunological roles of stearoyl-CoA desaturase 1 (SCD1), which is essential for fatty acid metabolism, in the cancer immune response. METHODS: We investigated the roles of SCD1 by inhibition with the chemical inhibitor or genetic manipulation in antitumor T cell responses and the therapeutic effect of anti-programmed cell death protein 1 (anti-PD-1) antibody using various mouse tumor models, and their cellular and molecular mechanisms. The roles of SCD1 in human cancers were also investigated by gene expression analyses of colon cancer tissues and by evaluating the related free fatty acids in sera obtained from patients with non-small cell lung cancer who were treated with anti-PD-1 antibody. RESULTS: Systemic administration of a SCD1 inhibitor in mouse tumor models enhanced production of CCL4 by cancer cells through reduction of Wnt/ß-catenin signaling and by CD8+ effector T cells through reduction of endoplasmic reticulum stress. It in turn promoted recruitment of dendritic cells (DCs) into the tumors and enhanced the subsequent induction and tumor accumulation of antitumor CD8+ T cells. SCD1 inhibitor was also found to directly stimulate DCs and CD8+ T cells. Administration of SCD1 inhibitor or SCD1 knockout in mice synergized with an anti-PD-1 antibody for its antitumor effects in mouse tumor models. High SCD1 expression was observed in one of the non-T cell-inflamed subtypes in human colon cancer, and serum SCD1 related fatty acids were correlated with response rates and prognosis of patients with non-small lung cancer following anti-PD-1 antibody treatment. CONCLUSIONS: SCD1 expressed in cancer cells and immune cells causes immunoresistant conditions, and its inhibition augments antitumor T cells and therapeutic effects of anti-PD-1 antibody. Therefore, SCD1 is an attractive target for the development of new diagnostic and therapeutic strategies to improve current cancer immunotherapies including immune checkpoint inhibitors.


Subject(s)
CD8-Positive T-Lymphocytes , Carcinoma, Non-Small-Cell Lung , Colonic Neoplasms , Lung Neoplasms , Stearoyl-CoA Desaturase , Animals , CD8-Positive T-Lymphocytes/immunology , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/immunology , Colonic Neoplasms/drug therapy , Colonic Neoplasms/immunology , Immune Checkpoint Inhibitors/pharmacology , Lung Neoplasms/drug therapy , Lung Neoplasms/immunology , Mice , Mice, Knockout , Stearoyl-CoA Desaturase/antagonists & inhibitors , Stearoyl-CoA Desaturase/immunology , Tumor Microenvironment , Wnt Signaling Pathway/immunology , beta Catenin/immunology
3.
J Pathol Clin Res ; 8(5): 458-469, 2022 09.
Article in English | MEDLINE | ID: mdl-35762092

ABSTRACT

Programmed cell death-1 (PD-1) and its ligand (PD-L1) are significant mediators of immune suppression in the tumor microenvironment. We focused on the immunological impact of PD-1/PD-L1 signaling during tumor progression in colorectal carcinoma (CRC) and its association with resistance to neoadjuvant chemoradiotherapy (NCRT) in locally advanced rectal carcinoma (LAd-RC). Histopathological and immunohistochemical analyses of 100 CRC cases (including 34 RC) without NCRT and 109 NCRT-treated LAd-RC cases were performed. Membranous tumoral PD-L1 expression was identified in 9 of 100 (9%) CRC cases, including 1 of 34 (2.9%) RC cases, but PD-L1 immunopositivity was not associated with any clinicopathological factors, with the exception of deficient mismatch repair (dMMR) status. In contrast, stromal PD-L1+ immune cells, which frequently exhibited coexpression of PD-1 and CD8 markers, were significantly correlated with tumor vessel invasion, nuclear ß-catenin+ tumor budding cancer stem cell (CSC)-like features, and unfavorable prognosis. In the LAd-RC cases, stromal CD8+ (but not PD-L1+) immune cell infiltration in pretreatment-biopsied samples was significantly and positively associated with therapeutic efficacy. After NCRT, tumoral PD-L1 expression was observed in only 2 of 83 (2.4%) tumors, independent of dMMR status, whereas high stromal PD-L1+ and tumoral nuclear ß-catenin positivity were significantly linked to a poor response to NCRT and high tumor budding features. In addition, high stromal PD-L1 immunoreactivity was significantly associated with poorer overall survival. In conclusion, a combination of stromal PD-L1+ immune cells and nuclear ß-catenin+ tumor budding may contribute to tumor progression in CRC and resistance to NCRT in LAd-RC, through formation of niche-like lesions that exhibit immune resistance and CSC properties.


Subject(s)
B7-H1 Antigen , Drug Resistance, Neoplasm , Programmed Cell Death 1 Receptor , Radiation Tolerance , Rectal Neoplasms , beta Catenin , B7-H1 Antigen/genetics , B7-H1 Antigen/immunology , Cell Nucleus/genetics , Cell Nucleus/immunology , Chemoradiotherapy, Adjuvant , Colorectal Neoplasms/genetics , Colorectal Neoplasms/immunology , Colorectal Neoplasms/pathology , Colorectal Neoplasms/therapy , Disease Progression , Drug Resistance, Neoplasm/genetics , Drug Resistance, Neoplasm/immunology , Gene Expression Regulation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic/immunology , Humans , Neoadjuvant Therapy , Prognosis , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/immunology , Radiation Tolerance/genetics , Radiation Tolerance/immunology , Rectal Neoplasms/genetics , Rectal Neoplasms/immunology , Rectal Neoplasms/pathology , Rectal Neoplasms/therapy , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology , beta Catenin/genetics , beta Catenin/immunology
4.
Cancer Lett ; 541: 215750, 2022 08 10.
Article in English | MEDLINE | ID: mdl-35609735

ABSTRACT

Immune checkpoint blockade (ICB) therapy is an important treatment option for individuals with cancer, but it has certain limitations. Identifying a better target that can overcome tumor immune escape and stimulate T cell activity is critical. This research aimed to delve into the molecular mechanism underlying the immunoregulatory function of metadherin (MTDH), which is a novel and potential therapeutic target in hepatocellular cancer (HCC). A small interfering RNA library was screened using the luciferase reporter assay and PD-L1 promoter. The Cancer Genome Atlas database and HCC tissues were used to investigate the relationship between MTDH and PD-L1. The association between MTDH and ß-catenin/lymphoid enhancer binding factor (LEF-1) was discovered by co-immunoprecipitation. The chromatin immunoprecipitation assay was used to investigate the interaction of MTDH with the PD-L1 promoter when LEF-1 expression was silenced. Locked nucleic acid antisense oligonucleotides (ASOs) were used to inhibit MTDH. We utilized in vitro co-cultures and in vivo syngeneic tumor development experiments to confirm the effectiveness of MTDH ASO combined with PD-1 monoclonal antibody (mAb). MTDH was demonstrated to be a PD-L1 modulator. MTDH increased PD-L1 expression and upregulated PD-L1 transcriptional activity through ß-catenin/LEF-1 signaling. More importantly, MTDH ASO improved the anti-PD-1 response and increased cytotoxic T-cell infiltration in PD-1 mAb-treated malignancies. MTDH effectively predicts the therapeutic efficacy of ICB therapy. Our results imply that combining MTDH ASO with PD-1 mAb could be a promising therapeutic strategy for HCC. In addition, MTDH is a potential novel biomarker for predicting the effectiveness of immune checkpoint inhibitor treatment.


Subject(s)
Antibodies, Monoclonal , B7-H1 Antigen , Carcinoma, Hepatocellular , Immune Checkpoint Inhibitors , Liver Neoplasms , Membrane Proteins , Oligonucleotides, Antisense , RNA-Binding Proteins , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/genetics , B7-H1 Antigen/immunology , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/immunology , Cell Line, Tumor , Humans , Immune Checkpoint Inhibitors/pharmacology , Liver Neoplasms/drug therapy , Liver Neoplasms/genetics , Liver Neoplasms/immunology , Membrane Proteins/genetics , Membrane Proteins/immunology , Oligonucleotides, Antisense/immunology , Programmed Cell Death 1 Receptor/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/immunology , Tumor Microenvironment , beta Catenin/genetics , beta Catenin/immunology
5.
Cell Rep ; 38(6): 110342, 2022 02 08.
Article in English | MEDLINE | ID: mdl-35139384

ABSTRACT

The mechanically activated Piezo channel plays a versatile role in conferring mechanosensitivity to various cell types. However, how it incorporates its intrinsic mechanosensitivity and cellular components to effectively sense long-range mechanical perturbation across a cell remains elusive. Here we show that Piezo channels are biochemically and functionally tethered to the actin cytoskeleton via the cadherin-ß-catenin mechanotransduction complex, whose perturbation significantly impairs Piezo-mediated responses. Mechanistically, the adhesive extracellular domain of E-cadherin interacts with the cap domain of Piezo1, which controls the transmembrane gate, while its cytosolic tail might interact with the cytosolic domains of Piezo1, which are in close proximity to its intracellular gates, allowing a direct focus of adhesion-cytoskeleton-transmitted force for gating. Specific disruption of the intermolecular interactions prevents cytoskeleton-dependent gating of Piezo1. Thus, we propose a force-from-filament model to complement the previously suggested force-from-lipids model for mechanogating of Piezo channels, enabling them to serve as versatile and tunable mechanotransducers.


Subject(s)
Actin Cytoskeleton/immunology , Cytoskeleton/metabolism , Ion Channels/metabolism , Mechanotransduction, Cellular/immunology , beta Catenin/metabolism , Actin Cytoskeleton/metabolism , Animals , Cadherins/immunology , Cadherins/metabolism , Humans , Ion Channel Gating , Mice , beta Catenin/immunology
6.
Front Immunol ; 12: 759565, 2021.
Article in English | MEDLINE | ID: mdl-34777372

ABSTRACT

Background: The emergence of immune checkpoint inhibitors (ICIs) marks the beginning of a new era of immunotherapy for hepatocellular carcinoma (HCC), however, not all patients respond successfully to this treatment. A major challenge for HCC immunotherapy is the development of ways to screen for those patients that would benefit from this type of treatment and determine the optimal treatment plan for individual patients. Therefore, it is important to find a biomarker which allows for the stratification of HCC patients, which distinguishes responders from non-responders, thereby further improving the clinical benefits for those undergoing immunotherapy. Methods: We used univariate and multivariate Cox risk proportional regression models to evaluate the relationship between non-synonymous mutations with a mutation frequency greater than 10%. We made a prognosis of an immunotherapy HCC cohort using mutation and prognosis data. An additional three HCC queues from the cbioportal webtool were used for further verification. The CIBERSORT, IPS, quanTIseq, and MCPcounter algorithms were used to evaluate the immune cells. PCA and z-score algorithm were used to calculate immune-related signature with published gene sets. Gene set enrichment analysis (GSEA) was used to compare the differences in the pathway-based enrichment scores of candidate genes between mutant and wild types. Results: Univariate and multivariate Cox results showed that only CTNNB1-Mutant(CTNNB1-MUT) was associated with progression-free survival (PFS) of HCC patients in the immunotherapy cohort. After excluding the potential bias introduced by other clinical features, it was found that CTNNB1-MUT served as an independent predictor of the prognosis of HCC patients after immunotherapy (P < 0.05; HR > 1). The results of the tumor immune microenvironment (TIME) analysis showed that patients with CTNNB1-MUT had significantly reduced activated immune cells [such as T cells, B cells, M1-type macrophages, and dendritic cells (DCs)], significantly increased M2-type macrophages, a significantly decreased expression of immunostimulating molecules, low activity of the immune activation pathways (cytokine pathway, immune cell activation and recruitment) and highly active immune depletion pathways (fatty acid metabolism, cholesterol metabolism, and Wnt pathway). Conclusions: In this study, we found CTNNB1-MUT to be a potential biomarker for HCC immunotherapy patients, because it identified those patients are less likely to benefit from ICIs.


Subject(s)
Biomarkers, Tumor/genetics , Carcinoma, Hepatocellular/diagnosis , Liver Neoplasms/diagnosis , beta Catenin/genetics , Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/immunology , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/therapy , Cohort Studies , Humans , Immunotherapy , Liver Neoplasms/immunology , Liver Neoplasms/therapy , Mutation , Prognosis , Proportional Hazards Models , beta Catenin/immunology
7.
mBio ; 12(6): e0282421, 2021 12 21.
Article in English | MEDLINE | ID: mdl-34781737

ABSTRACT

The signaling pathways activated following interaction between dendritic cells (DCs) and a pathogen determine the polarization of effector T-cell and regulatory T-cell (Treg) responses to the infection. Several recent studies, mostly in the context of bacterial infections, have shown that the Wnt/ß-catenin pathway plays a major role in imparting tolerogenic features in DCs and in promotion of Treg responses. However, the significance of the Wnt/ß-catenin pathway's involvement in regulating the immune response to the fungal species is not known. Using Aspergillus fumigatus, a ubiquitous airborne opportunistic fungal species, we show here that fungi activate the Wnt/ß-catenin pathway in human DCs and are critical for mediating the immunosuppressive Treg responses. Pharmacological inhibition of this pathway in DCs led to inhibition of maturation-associated molecules and interleukin 10 (IL-10) secretion without affecting the majority of the inflammatory cytokines. Furthermore, blockade of Wnt signaling in DCs suppressed DC-mediated Treg responses in CD4+ T cells and downregulated both tumor necrosis factor alpha (TNF-α) and IL-10 responses in CD8+ T cells. Mechanistically, induction of ß-catenin pathway by A. fumigatus required C-type lectin receptors and promoted Treg polarization via the induction of programmed death-ligand 1 on DCs. Further investigation on the identity of fungal molecular patterns has revealed that the cell wall polysaccharides ß-(1, 3)-glucan and α-(1, 3)-glucan, but not chitin, possess the capacity to activate the ß-catenin pathway. Our data suggest that the Wnt/ß-catenin pathway is a potential therapeutic target to selectively suppress the Treg response and to sustain the protective Th1 response in the context of invasive aspergillosis caused by A. fumigatus. IMPORTANCE The balance between effector CD4+ T-cell and immunosuppressive regulatory T-cell (Treg) responses determines the outcome of an infectious disease. The signaling pathways that regulate human CD4+ T-effector versus Treg responses to the fungi are not completely understood. By using Aspergillus fumigatus, a ubiquitous opportunistic fungal species, we show that fungi activate the Wnt/ß-catenin pathway in human dendritic cells (DCs) that promotes Treg responses via induction of immune checkpoint molecule programmed death ligand 1 on DCs. Blockade of the Wnt/ß-catenin pathway in DCs led to the selective inhibition of Treg without affecting the Th1 response. Dissection of the identity of A. fumigatus pathogen-associated molecular patterns (PAMPs) revealed that cell wall polysaccharides exhibit selectivity in their capacity to activate the ß-catenin pathway in DCs. Our data thus provide a pointer that Wnt/ß-catenin pathway represents potential therapeutic target to selectively suppress Treg responses and to sustain protective a Th1 response against invasive fungal diseases.


Subject(s)
Aspergillosis/immunology , Aspergillus fumigatus/physiology , B7-H1 Antigen/immunology , Dendritic Cells/immunology , T-Lymphocytes, Regulatory/immunology , beta Catenin/immunology , Aspergillosis/genetics , Aspergillosis/microbiology , B7-H1 Antigen/genetics , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Humans , Interleukin-10/genetics , Interleukin-10/immunology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology , Wnt Signaling Pathway , beta Catenin/genetics
8.
Pathobiology ; 88(5): 351-358, 2021.
Article in English | MEDLINE | ID: mdl-34237735

ABSTRACT

INTRODUCTION: An immunohistochemical study has occasionally been performed to diagnose anaplastic thyroid carcinoma (ATC). However, antibodies to confirm the undifferentiated nature of ATC have not yet been evaluated. The aim of this study was to evaluate E-cadherin and ß-catenin expressions in immunoreactivity to determine undifferentiated carcinoma cells in the diagnosis of ATC. METHODS: We immunohistochemically examined 29 ATCs, 30 poorly differentiated thyroid carcinomas (PDTCs), 22 well-differentiated thyroid carcinomas (WDTCs), and 3 squamous cell carcinomas. Antibodies for thyroid transcription factor-1 (TTF-1), paired-box gene 8 (PAX8), ß-catenin, and E-cadherin were used. RESULTS: All WDTCs tested positive for TTF-1, PAX8, and E-cadherin. The positive rates of TTF-1, PAX8, and E-cadherin were 93.3, 93.3, and 100%, respectively, in PDTCs and 17.2, 51.7, and 10.3%, respectively, in ATCs. WDTC expressed the lateral cell membrane staining for ß-catenin and E-cadherin, whereas PDTC showed circumferential cell membranous expression (fishnet pattern). ß-catenin cell membrane expression in ATCs is lost or discontinuous. Carcinoma cells with ß-catenin nuclear expression without cell membranous expression were scattered in 72.4% of ATCs but were not observed in the other carcinomas. CONCLUSION: We propose 3 immunohistochemical findings to determine undifferentiated carcinoma cells in the diagnosis of ATC: (1) ß-catenin nuclear expression with no or reduced cell membranous expression, (2) the loss or discontinuous pattern of E-cadherin expression, and (3) the loss of PAX8 nuclear expression.


Subject(s)
Cadherins/genetics , Carcinoma, Squamous Cell/genetics , Immunohistochemistry/methods , Thyroid Carcinoma, Anaplastic/genetics , beta Catenin/genetics , Biomarkers, Tumor , Cadherins/immunology , Carcinoma, Squamous Cell/immunology , Carcinoma, Squamous Cell/metabolism , Humans , Immunohistochemistry/standards , Paraffin Embedding , Thyroid Carcinoma, Anaplastic/immunology , Thyroid Gland/pathology , beta Catenin/immunology
9.
Fish Shellfish Immunol ; 118: 228-240, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34284111

ABSTRACT

ß-catenin is a structural protein that makes the cell-cell connection in adherence junctions. Besides the structural functions, it also plays a role as a central transducer of the canonical Wnt signaling cascade, regulating nearly four hundred genes related to various cellular processes. Recently the immune functions of ß-catenin during pathogenic invasion have gained more attention. In the present study, we elucidated the immune function of fish ß-catenin by identifying and characterizing the ß-catenin homolog (PhCatß) from redlip mullet, Planiliza haematocheila. The complete open reading frame of PhCatß consists of 2352 bp, which encodes a putative ß-catenin homolog (molecular weight: 85.7 kDa). Multiple sequence alignment analysis revealed that ß-catenin is highly conserved in vertebrates. Phylogenetic reconstruction demonstrated the close evolutionary relationship between PhCatß and other fish ß-catenin counterparts. The tissue distribution analysis showed the highest mRNA expression of PhCatß in heart tissues of the redlip mullet under normal physiological conditions. While in response to pathogenic stress, the PhCatß transcription level was dramatically increased in the spleen and gill tissues. The overexpression of PhCatß stimulated M2 polarization and cell proliferation of murine RAW 264.7 macrophage. In fish cells, the overexpression of PhCatß resulted in a significant upregulation of antiviral gene transcription and vice versa. Moreover, the overexpression of PhCatß could inhibit the replication of VHSV in FHM cells. Our results strongly suggest that PhCatß plays a role in macrophage activation and antiviral immune response in redlip mullet.


Subject(s)
Antiviral Agents , Cytosol , Fish Proteins , Macrophage Activation , Smegmamorpha , beta Catenin , Animals , Antiviral Agents/chemistry , Antiviral Agents/immunology , Antiviral Agents/metabolism , Evolution, Molecular , Fish Proteins/chemistry , Fish Proteins/genetics , Fish Proteins/immunology , Fish Proteins/metabolism , Gene Expression Profiling , Macrophage Activation/drug effects , Macrophages/drug effects , Mice , Organ Specificity , Phylogeny , RAW 264.7 Cells , Smegmamorpha/classification , Smegmamorpha/genetics , beta Catenin/chemistry , beta Catenin/genetics , beta Catenin/immunology , beta Catenin/metabolism
10.
Mech Ageing Dev ; 197: 111517, 2021 07.
Article in English | MEDLINE | ID: mdl-34139213

ABSTRACT

Reversible cellular senescence was demonstrated previously to constitute colon cancer cell response to methotrexate. The current study presents a comparison of two senescent states of colon cancer cells, arrested and reversing, resulting from respectively, 120 h exposure to the drug, and 48 h exposure followed by 96 h regrowth in drug-free media. The upregulation of immunoproteasome subunit-coding genes and the increase in human leukocyte antigen HLA-A/B/C membrane level indicated MHC-I-restricted antigen presentation as common to both senescent states. Nuclear factor NF-κB p65 level decreased and activating protein AP-1: c-Jun, Fra2 and JunB nuclear levels increased in both senescent cell populations. Notably, the increase in AP-1- dependent transcription occurred after 48 h exposure to methotrexate. ß-catenin nuclear level increased after 48 h exposure to the drug and remained as such only in senescence-arrested cells. ß-catenin level was found uncoupled from the protein phosphorylation status indicating the deregulation of ß-catenin signaling in colon cancer cells employed in the study. These findings carry implications for both, a general mechanism of senescence establishment and putative advantages for colon cancer treatment.


Subject(s)
Antigen Presentation , Cellular Senescence/drug effects , Colonic Neoplasms/immunology , Methotrexate/pharmacology , Neoplasm Proteins/immunology , Signal Transduction/immunology , Transcription Factor AP-1/immunology , beta Catenin/immunology , Cell Line, Tumor , Cellular Senescence/immunology , Humans
11.
Pathol Int ; 71(6): 392-399, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33788979

ABSTRACT

Desmoid-type fibromatosis (DF) is a locally aggressive but non-metastatic (myo)fibroblastic neoplasm. A hallmark of the tumor is nuclear positivity for beta-catenin in immunohistochemistry due mostly to CTNNB1 mutations. However, a recent study has reported that even beta-catenin 'nuclear-negative' DFs can harbor CTNNB1 mutations and that the positive ratio of nuclear beta-catenin in DF is different among antibodies. Here, we reviewed soft tissue lesions for which the possibility of DF was considered and compared the sensitivity and specificity of nuclear beta-catenin for the diagnosis of DF among commonly used anti-beta-catenin antibodies, i.e., clone beta-catenin 1, 17C2 and 14. We analyzed 26 cases of DF, 28 cases of benign fibroblastic lesions, and 27 cases of other soft tissue tumors. The sensitivity and specificity of nuclear beta-catenin for the diagnosis of DF were different among antibodies; 54% and 98% in clone beta-catenin 1, 85% and 84% in 17C2, and 96% and 62% in 14. IHC of LEF1 showed comparable results with IHC of beta-catenin, with a sensitivity of 88% and specificity of 76%. Additionally, when beta-catenin 1 was used, DFs showed characteristic dotted cytoplasmic staining, often appearing as rings. Our results might be helpful for making a correct diagnosis of DF.


Subject(s)
Antibodies, Neoplasm , Fibromatosis, Aggressive , beta Catenin , Adolescent , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor , Child , Child, Preschool , Diagnosis, Differential , Female , Fibromatosis, Aggressive/diagnosis , Fibromatosis, Aggressive/pathology , Humans , Immunohistochemistry , Infant , Male , Middle Aged , Mutation , Soft Tissue Neoplasms/diagnosis , Soft Tissue Neoplasms/pathology , Young Adult , beta Catenin/genetics , beta Catenin/immunology , beta Catenin/metabolism
12.
BMC Complement Med Ther ; 21(1): 43, 2021 Jan 23.
Article in English | MEDLINE | ID: mdl-33485352

ABSTRACT

BACKGROUND: Periodontitis is a chronic infection initiated by oral bacterial and their virulence factors, yet the severity of periodontitis is largely determined by the dysregulated host immuno-inflammatory response. Baicalein is a flavonoid extracted from Scutellaria baicalensis with promising anti-inflammatory properties. This study aims to clarify the anti-inflammatory and osteogenic effects of baicalein in periodontal ligament cells (PDLCs) treated with lipopolysaccharides (LPS). METHODS: Human PDLCs were incubated with baicalein (0-100 µM) for 2 h prior to LPS challenge for 24 h. MTT analysis was adopted to assess the cytoxicity of baicalein. The mRNA and protein expression of inflammatory and osteogenic markers were measured by real-time polymerase chain reaction (PCR), western blot and enzyme-linked immunosorbent assay (ELISA) as appropriate. Alkaline phosphatase (ALP) and Alizarin red S (ARS) staining were performed to evaluate the osteogenic differentiation of PDLCs. The expression of Wnt/ß-catenin and mitogen-activated protein kinase (MAPK) signaling related proteins was assessed by western blot. RESULTS: MTT results showed that baicalein up to 100 µM had no cytotoxicity on PDLCs. Baicalein significantly attenuated the inflammatory factors induced by LPS, including interleukin-1ß (IL-1ß), tumor necrosis factor-α (TNF-α), matrix metalloprotein-1 (MMP-1), MMP-2 and monocyte chemoattractant protein 1 (MCP-1) at both mRNA and protein level. Moreover, MAPK signaling (ERK, JNK and p38) was significantly inhibited by baicalein, which may account for the mitigated inflammatory response. Next, we found that baicalein effectively restored the osteogenic differentiation of LPS-treated PDLCs, as shown by the increased ALP and ARS staining. Accordingly, the protein and gene expression of osteogenic markers, namely runt-related transcription factor 2 (RUNX2), collagen-I, and osterix were markedly upregulated. Importantly, baicalein could function as the Wnt/ß-catenin signaling activator, which may lead to the increased osteoblastic differentiation of PDLCs. CONCLUSIONS: With the limitation of the study, we provide in vitro evidence that baicalein ameliorates inflammatory response and restores osteogenesis in PDLCs challenged with LPS, indicating its potential use as the host response modulator for the management of periodontitis.


Subject(s)
Drugs, Chinese Herbal/pharmacology , Flavanones/pharmacology , Osteogenesis/drug effects , Periodontal Ligament/drug effects , Periodontitis/drug therapy , Scutellaria baicalensis/chemistry , Alkaline Phosphatase/genetics , Alkaline Phosphatase/immunology , Cell Differentiation/drug effects , Cells, Cultured , Chemokine CCL2/genetics , Chemokine CCL2/immunology , Humans , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Lipopolysaccharides/adverse effects , Matrix Metalloproteinase 1/genetics , Matrix Metalloproteinase 1/immunology , Periodontal Ligament/cytology , Periodontal Ligament/immunology , Periodontitis/genetics , Periodontitis/immunology , Periodontitis/physiopathology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology , Wnt Signaling Pathway/drug effects , beta Catenin/genetics , beta Catenin/immunology
13.
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
14.
Med Sci Monit ; 26: e925494, 2020 Sep 18.
Article in English | MEDLINE | ID: mdl-32945289

ABSTRACT

BACKGROUND Immunotherapy is one of the research hotspots in the field of hepatocellular carcinoma (HCC). Successive clinical trials have shown that patients with CTNNB1 mutations are resistant to immunotherapy, but the mechanism is still unclear. MATERIAL AND METHODS We identified differentially expressed immune genes (DEIGs) in patients with and without CTNNB1 mutations in the Cancer Genome Atlas (TCGA) database and then paired them to explore any correlation with prognosis. Univariate Cox regression analysis and Lasso regression analysis were used to develop the prognostic model. We first divided the TCGA cohort into 29 subgroups for internal validation and then used the International Cancer Genome Consortium (ICGC) cohort to conduct external validation. We also used a CIBERSORT algorithm to quantify immune infiltration of the different risk groups. RESULTS The novel prognostic model consisted of 45 immune-gene pairs with general applicability. It was more accurate than the traditional prognostic signature, which is based on gene expression by comparison of area under the receiver operating characteristic curve (AUC) values. The infiltration proportion of B cells, CD8 T lymphocytes, activated natural killer cells, and M1 macrophages in the low-risk group was greater in the high-risk group, while the infiltration proportion of M0 and M2 macrophages was greater in the high-risk group. CONCLUSIONS In this study, a novel approach was proposed for evaluating HCC prognosis, which may be useful in evaluatingthe intensity of the immune response in the HCC microenvironment.


Subject(s)
Carcinoma, Hepatocellular , Databases, Nucleic Acid , Liver Neoplasms , Models, Immunological , Neoplasm Proteins , beta Catenin , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/immunology , Female , Humans , Liver Neoplasms/genetics , Liver Neoplasms/immunology , Macrophages/immunology , Male , Neoplasm Proteins/genetics , Neoplasm Proteins/immunology , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology , beta Catenin/genetics , beta Catenin/immunology
15.
Cancer Res ; 80(10): 2004-2016, 2020 05 15.
Article in English | MEDLINE | ID: mdl-32156780

ABSTRACT

Dysregulation of Wnt/ß-catenin signaling is frequently observed in human gastric cancer. Elucidation of the tumor immune microenvironment is essential for understanding tumorigenesis and for the development of immunotherapeutic strategies. However, it remains unclear how ß-catenin signaling regulates the tumor immune microenvironment in the stomach. Here, we identify CCL28 as a direct transcriptional target gene of ß-catenin/T-cell factor (TCF). Protein levels of ß-catenin and CCL28 positively correlated in human gastric adenocarcinoma. ß-Catenin-activated CCL28 recruited regulatory T (Treg) cells in a transwell migration assay. In a clinically relevant mouse gastric cancer model established by Helicobacter (H.) felis infection and N-methyl-N-nitrosourea (MNU) treatment, inhibition of ß-catenin/TCF activity by a pharmacologic inhibitor iCRT14 suppressed CCL28 expression and Treg cell infiltration in the stomach. Moreover, an anti-CCL28 antibody attenuated Treg cell infiltration and tumor progression in H. felis/MNU mouse models. Diphtheria toxin-induced Treg cell ablation restrained gastric cancer progression in H. felis/MNU-treated DEREG (Foxp3-DTR) mice, clarifying the tumor-promoting role of Treg cells. Thus, the ß-catenin-CCL28-Treg cell axis may serve as an important mechanism for immunosuppression of the stomach tumor microenvironment. Our findings reveal an immunoregulatory role of ß-catenin signaling in stomach tumors and highlight the therapeutic potential of CCL28 blockade for the treatment of gastric cancer. SIGNIFICANCE: These findings demonstrate an immunosuppressive role of tumor-intrinsic ß-catenin signaling and the therapeutic potential of CCL28 blockade in gastric cancer.


Subject(s)
Adenocarcinoma/pathology , Chemokines, CC/metabolism , Stomach Neoplasms/pathology , T-Lymphocytes, Regulatory/immunology , Tumor Microenvironment/immunology , beta Catenin/metabolism , Adenocarcinoma/immunology , Animals , Chemokines, CC/immunology , Disease Progression , Gene Expression Regulation, Neoplastic/physiology , Humans , Lymphocytes, Tumor-Infiltrating/immunology , Mice , Stomach Neoplasms/immunology , Tumor Escape/physiology , beta Catenin/immunology
16.
PLoS One ; 15(3): e0230563, 2020.
Article in English | MEDLINE | ID: mdl-32210470

ABSTRACT

Despite antiretroviral therapy human immunodeficiency virus type-1 (HIV-1) infection results in neuroinflammation of the central nervous system that can cause HIV-associated neurocognitive disorders (HAND). The molecular mechanisms involved in the development of HAND are unclear, however, they are likely due to both direct and indirect consequences of HIV-1 infection and inflammation of the central nervous system. Additionally, opioid abuse in infected individuals has the potential to exacerbate HIV-comorbidities, such as HAND. Although restricted for productive HIV replication, astrocytes (comprising 40-70% of all brain cells) likely play a significant role in neuropathogenesis in infected individuals due to the production and response of viral proteins. The HIV-1 protein Tat is critical for viral transcription, causes neuroinflammation, and can be secreted from infected cells to affect uninfected bystander cells. The Wnt/ß-catenin signaling cascade plays an integral role in restricting HIV-1 infection in part by negatively regulating HIV-1 Tat function. Conversely, Tat can overcome this negative regulation and inhibit ß-catenin signaling by sequestering the critical transcription factor TCF-4 from binding to ß-catenin. Here, we aimed to explore how opiate exposure affects Tat-mediated suppression of ß-catenin in astrocytes and the downstream modulation of neuroinflammatory genes. We observed that morphine can potentiate Tat suppression of ß-catenin activity in human astrocytes. In contrast, Tat mutants deficient in secretion, and lacking neurotoxic effects, do not affect ß-catenin activity in the presence or absence of morphine. Finally, morphine treatment of astrocytes was sufficient to reduce the expression of genes involved in neuroinflammation. Examining the molecular mechanisms of how HIV-1 infection and opiate exposure exacerbate neuroinflammation may help us inform or predict disease progression prior to HAND development.


Subject(s)
Analgesics, Opioid/adverse effects , HIV Infections/complications , HIV-1/drug effects , Morphine/adverse effects , Neurocognitive Disorders/etiology , Substance-Related Disorders/complications , tat Gene Products, Human Immunodeficiency Virus/immunology , Astrocytes/drug effects , Astrocytes/immunology , Astrocytes/virology , Cell Line , Cells, Cultured , HIV Infections/immunology , HIV Infections/virology , HIV-1/immunology , Humans , Neurocognitive Disorders/immunology , Neurocognitive Disorders/virology , Substance-Related Disorders/immunology , beta Catenin/immunology
17.
Front Immunol ; 11: 122, 2020.
Article in English | MEDLINE | ID: mdl-32132993

ABSTRACT

Dendritic cells (DCs) control the strength and quality of antigen-specific adaptive immune responses. This is critical for launching a robust immunity against invading pathogens while maintaining a state of tolerance to self-antigens. However, this also represents a fundamental barrier to anti-tumor immune responses and cancer immunotherapy. DCs in the tumor microenvironment (TME) play a key role in this process. The factors in the TME and signaling networks that program DCs to a regulatory state are not fully understood. Recent advances point to novel mechanisms by which the canonical Wnt signaling cascade in DCs regulates immune suppression, and the same pathway in tumors is associated with the evasion of anti-tumor immunity. Here, we review these recent advances in the context of the pleiotropic effects of the Wnts in shaping anti-tumor immune responses by modulating DC functions. In addition, we will discuss how Wnt/ß-catenin pathway in DCs can be targeted for successful cancer immunotherapy.


Subject(s)
Dendritic Cells/immunology , Dendritic Cells/metabolism , Immunity/immunology , Tumor Microenvironment/immunology , Tumor Microenvironment/physiology , Wnt Signaling Pathway/immunology , Humans , Immunotherapy , Neoplasms/immunology , Signal Transduction/immunology , beta Catenin/immunology , beta Catenin/metabolism
18.
Nat Commun ; 11(1): 821, 2020 02 10.
Article in English | MEDLINE | ID: mdl-32041953

ABSTRACT

The diversity of the naïve T cell repertoire drives the replenishment potential and capacity of memory T cells to respond to immune challenges. Attrition of the immune system is associated with an increased prevalence of pathologies in aged individuals, but whether stem cell memory T lymphocytes (TSCM) contribute to such attrition is still unclear. Using single cells RNA sequencing and high-dimensional flow cytometry, we demonstrate that TSCM heterogeneity results from differential engagement of Wnt signaling. In humans, aging is associated with the coupled loss of Wnt/ß-catenin signature in CD4 TSCM and systemic increase in the levels of Dickkopf-related protein 1, a natural inhibitor of the Wnt/ß-catenin pathway. Functional assays support recent thymic emigrants as the precursors of CD4 TSCM. Our data thus hint that reversing TSCM defects by metabolic targeting of the Wnt/ß-catenin pathway may be a viable approach to restore and preserve immune homeostasis in the context of immunological history.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Precursor Cells, T-Lymphoid/immunology , Wnt Signaling Pathway/immunology , Aging/immunology , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Gene Expression Profiling , HIV Infections/immunology , Humans , Immunologic Memory , Intercellular Signaling Peptides and Proteins/metabolism , Mice , Thymus Gland/immunology , Wnt Signaling Pathway/genetics , beta Catenin/immunology
19.
Front Immunol ; 10: 2777, 2019.
Article in English | MEDLINE | ID: mdl-31849960

ABSTRACT

Mast cells constitutively express ß-catenin and expand in solid tumors such as colon and skin cancer. However, the role of ß-catenin signaling in mast cells and the cause or effect of mast cell expansion and tumor growth has yet to be established. In earlier studies we used mast cell depletion and protease staining approaches, to provide evidence for a causative role of mast cells in small bowel polyposis, and related specific phenotypes and distributions of tumor infiltrating mast cells to stages of tumor growth. Here we report that, stabilization of ß-catenin expands mast cells to promote high incidence of colon polyposis and infrequent small bowel polyps and skin cancer. Expression of a dominant acting ß-catenin in mast cells (5CreCAT) stimulated maturation and expression of granule stored proteases. Both mucosal and connective tissue type mast cells accumulated in colonic small bowel polyps independent of gender, and mice developed chronic systemic inflammation with splenomegaly. Reconstitution of polyposis-prone mice with bone marrow from 5CreCAT mice resulted in focal expansion of connective tissue like mast cells, which are normally rare in benign polyps and characteristically expand during adenoma-to-carcinoma transition. Our findings highlight a hitherto unknown contribution of ß-catenin signaling in mast cells to their maturation and to increased risk of colon cancer.


Subject(s)
Colonic Neoplasms/immunology , Mast Cells/immunology , beta Catenin/immunology , Animals , Bone Marrow , Cell Proliferation , Cells, Cultured , Colon/pathology , Colonic Neoplasms/pathology , Connective Tissue , Female , Inflammation/immunology , Male , Mice , Signal Transduction
20.
J Immunother Cancer ; 7(1): 308, 2019 11 15.
Article in English | MEDLINE | ID: mdl-31730502

ABSTRACT

BACKGROUND: Clinical response to MAPK inhibitors in metastatic melanoma patients is heterogeneous for reasons still needing to be elucidated. As the patient immune activity contributes to treatment clinical benefit, the pre-existing level of immunity at tumor site may provide biomarkers of disease outcome to therapy. Here we investigated whether assessing the density and spatial tissue distribution of key immune cells in the tumor microenvironment could identify patients predisposed to respond to MAPK inhibitors. METHODS: Pretreatment tumor biopsies from a total of 213 patients (158 for the training set and 55 for the validation set) treated with BRAF or BRAF/MEK inhibitors within the Italian Melanoma Intergroup were stained with selected immune markers (CD8, CD163, ß-catenin, PD-L1, PD-L2). Results, obtained by blinded immunohistochemical scoring and digital image analysis, were correlated with clinical response and outcome by multivariate logistic models on response to treatment and clinical outcome, adjusted for American Joint Committee on Cancer stage, performance status, lactate dehydrogenase and treatment received. RESULTS: Patients with high intratumoral, but not peritumoral, CD8+ T cells and concomitantly low CD163+ myeloid cells displayed higher probability of response (OR 9.91, 95% CI 2.23-44.0, p = 0.003) and longer overall survival (HR 0.34, 95% CI 0.16-0.72, p = 0.005) compared to those with intratumoral low CD8+ T cells and high CD163+ myeloid cells. The latter phenotype was instead associated with a shorter progression free survival (p = 0.010). In contrast, PD-L1 and PD-L2 did not correlate with clinical outcome while tumor ß-catenin overexpression showed association with lower probability of response (OR 0.48, 95% CI 0.21-1.06, p = 0.068). CONCLUSIONS: Analysis of the spatially constrained distribution of CD8+ and CD163+ cells, representative of the opposite circuits of antitumor vs protumor immunity, respectively, may assist in identifying melanoma patients with improved response and better outcome upon treatment with MAPK inhibitors. These data underline the role of endogenous immune microenvironment in predisposing metastatic melanoma patients to benefit from therapies targeting driver-oncogenic pathways.


Subject(s)
MAP Kinase Kinase Kinases/antagonists & inhibitors , Melanoma/drug therapy , Melanoma/immunology , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Aged , Antigens, CD/immunology , Antigens, Differentiation, Myelomonocytic/immunology , B7-H1 Antigen/immunology , CD8 Antigens/immunology , Female , Humans , Kaplan-Meier Estimate , Male , Melanoma/mortality , Middle Aged , Programmed Cell Death 1 Ligand 2 Protein/immunology , Receptors, Cell Surface/immunology , Tumor Microenvironment/immunology , beta Catenin/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...