Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Cancers (Basel) ; 14(6)2022 Mar 21.
Article in English | MEDLINE | ID: mdl-35326747

ABSTRACT

Protein kinase C (PKC)-θ is a serine/threonine kinase with both cytoplasmic and nuclear functions. Nuclear chromatin-associated PKC-θ (nPKC-θ) is increasingly recognized to be pathogenic in cancer, whereas its cytoplasmic signaling is restricted to normal T-cell function. Here we show that nPKC-θ is enriched in circulating tumor cells (CTCs) in patients with triple-negative breast cancer (TNBC) brain metastases and immunotherapy-resistant metastatic melanoma and is associated with poor survival in immunotherapy-resistant disease. To target nPKC-θ, we designed a novel PKC-θ peptide inhibitor (nPKC-θi2) that selectively inhibits nPKC-θ nuclear translocation but not PKC-θ signaling in healthy T cells. Targeting nPKC-θ reduced mesenchymal cancer stem cell signatures in immunotherapy-resistant CTCs and TNBC xenografts. PKC-θ was also enriched in the nuclei of CD8+ T cells isolated from stage IV immunotherapy-resistant metastatic cancer patients. We show for the first time that nPKC-θ complexes with ZEB1, a key repressive transcription factor in epithelial-to-mesenchymal transition (EMT), in immunotherapy-resistant dysfunctional PD1+/CD8+ T cells. nPKC-θi2 inhibited the ZEB1/PKC-θ repressive complex to induce cytokine production in CD8+ T cells isolated from patients with immunotherapy-resistant disease. These data establish for the first time that nPKC-θ mediates immunotherapy resistance via its activity in CTCs and dysfunctional CD8+ T cells. Disrupting nPKC-θ but retaining its cytoplasmic function may offer a means to target metastases in combination with chemotherapy or immunotherapy.

2.
Front Immunol ; 11: 1228, 2020.
Article in English | MEDLINE | ID: mdl-32612611

ABSTRACT

Lysine specific demethylase 1 (LSD1) is a key epigenetic eraser enzyme implicated in cancer metastases and recurrence. Nuclear LSD1 phosphorylated at serine 111 (nLSD1p) has been shown to be critical for the development of breast cancer stem cells. Here we show that circulating tumor cells isolated from immunotherapy-resistant metastatic melanoma patients express higher levels of nLSD1p compared to responders, which is associated with co-expression of stem-like, mesenchymal genes. Targeting nLSD1p with selective nLSD1 inhibitors better inhibits the stem-like mesenchymal signature than traditional FAD-specific LSD1 catalytic inhibitors such as GSK2879552. We also demonstrate that nLSD1p is enriched in PD-1+CD8+ T cells from resistant melanoma patients and 4T1 immunotherapy-resistant mice. Targeting the LSD1p nuclear axis induces IFN-γ/TNF-α-expressing CD8+ T cell infiltration into the tumors of 4T1 immunotherapy-resistant mice, which is further augmented by combined immunotherapy. Underpinning these observations, nLSD1p is regulated by the key T cell exhaustion transcription factor EOMES in dysfunctional CD8+ T cells. EOMES co-exists with nLSD1p in PD-1+CD8+ T cells in resistant patients, and nLSD1p regulates EOMES nuclear dynamics via demethylation/acetylation switching of critical EOMES residues. Using novel antibodies to target these post-translational modifications, we show that EOMES demethylation/acetylation is reciprocally expressed in resistant and responder patients. Overall, we show for the first time that dual inhibition of metastatic cancer cells and re-invigoration of the immune system requires LSD1 inhibitors that target the nLSD1p axis.


Subject(s)
Cellular Reprogramming/drug effects , Cellular Reprogramming/genetics , Histone Demethylases/genetics , Neoplasms/etiology , T-Box Domain Proteins/metabolism , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , Animals , Biomarkers , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Cell Nucleus/genetics , Cell Nucleus/metabolism , Female , Flow Cytometry , Fluorescent Antibody Technique , Gene Expression , Gene Expression Profiling , High-Throughput Nucleotide Sequencing , Histone Demethylases/metabolism , Humans , Immunotherapy , Mice , Neoplasms/metabolism , Neoplasms/pathology , Neoplasms/therapy , T-Box Domain Proteins/genetics , T-Lymphocytes/immunology , Treatment Outcome
3.
Int J Mol Sci ; 20(12)2019 Jun 24.
Article in English | MEDLINE | ID: mdl-31238530

ABSTRACT

DUSP6 is a dual-specificity phosphatase (DUSP) involved in breast cancer progression, recurrence, and metastasis. DUSP6 is predominantly cytoplasmic in HER2+ primary breast cancer cells, but the expression and subcellular localization of DUSPs, especially DUSP6, in HER2-positive circulating tumor cells (CTCs) is unknown. Here we used the DEPArray system to identify and isolate CTCs from metastatic triple negative breast cancer (TNBC) patients and performed single-cell NanoString analysis to quantify cancer pathway gene expression in HER2-positive and HER2-negative CTC populations. All TNBC patients contained HER2-positive CTCs. HER2-positive CTCs were associated with increased ERK1/ERK2 expression, which are direct DUSP6 targets. DUSP6 protein expression was predominantly nuclear in breast CTCs and the brain metastases but not pleura or lung metastases of TNBC patients. Therefore, nuclear DUSP6 may play a role in the association with cancer spreading in TNBC patients, including brain metastasis.


Subject(s)
Biomarkers, Tumor , Brain Neoplasms/secondary , Dual Specificity Phosphatase 6/genetics , Gene Expression Regulation, Neoplastic , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology , ATP Binding Cassette Transporter, Subfamily B/genetics , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents, Immunological/pharmacology , Cell Line, Tumor , Cell Nucleus/genetics , Disease Models, Animal , Dual Specificity Phosphatase 6/antagonists & inhibitors , Dual Specificity Phosphatase 6/metabolism , Female , Fluorescent Antibody Technique , Gene Expression Regulation, Neoplastic/drug effects , Histones/metabolism , Humans , MAP Kinase Signaling System , Mice , Neoplasm Invasiveness , Neoplasm Staging , Neoplastic Cells, Circulating/metabolism , Neoplastic Cells, Circulating/pathology , Protein Binding , Protein Transport , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Single-Cell Analysis , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/therapy , Xenograft Model Antitumor Assays , p300-CBP Transcription Factors/metabolism
4.
Cell Mol Gastroenterol Hepatol ; 7(4): 819-839, 2019.
Article in English | MEDLINE | ID: mdl-30831321

ABSTRACT

BACKGROUND & AIMS: The early events by which inflammation promotes cancer are still not fully defined. The MCC gene is silenced by promoter methylation in colitis-associated and sporadic colon tumors, but its functional significance in precancerous lesions or polyps is not known. Here, we aimed to determine the impact of Mcc deletion on the cellular pathways and carcinogenesis associated with inflammation in the mouse proximal colon. METHODS: We generated knockout mice with deletion of Mcc in the colonic/intestinal epithelial cells (MccΔIEC) or in the whole body (MccΔ/Δ). Drug-induced lesions were analyzed by transcriptome profiling (at 10 weeks) and histopathology (at 20 weeks). Cell-cycle phases and DNA damage proteins were analyzed by flow cytometry and Western blot of hydrogen peroxide-treated mouse embryo fibroblasts. RESULTS: Transcriptome profiling of the lesions showed a strong response to colon barrier destruction, such as up-regulation of key inflammation and cancer-associated genes as well as 28 interferon γ-induced guanosine triphosphatase genes, including the homologs of Crohn's disease susceptibility gene IRGM. These features were shared by both Mcc-expressing and Mcc-deficient mice and many of the altered gene expression pathways were similar to the mesenchymal colorectal cancer subtype known as consensus molecular subtype 4 (CMS4). However, Mcc deletion was required for increased carcinogenesis in the lesions, with adenocarcinoma in 59% of MccΔIEC compared with 19% of Mcc-expressing mice (P = .002). This was not accompanied by hyperactivation of ß-catenin, but Mcc deletion caused down-regulation of DNA repair genes and a disruption of DNA damage signaling. CONCLUSIONS: Loss of Mcc may promote cancer through a failure to repair inflammation-induced DNA damage. We provide a comprehensive transcriptome data set of early colorectal lesions and evidence for the in vivo significance of MCC silencing in colorectal cancer.


Subject(s)
Colorectal Neoplasms/genetics , Gene Deletion , Genes, MCC , Inflammation/genetics , Animals , Cadherins/metabolism , Colon/drug effects , Colon/pathology , Colorectal Neoplasms/pathology , DNA Repair/genetics , Disease Models, Animal , Down-Regulation/drug effects , Down-Regulation/genetics , Female , GTP Phosphohydrolases/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Inflammation/pathology , Interferon-gamma/pharmacology , Male , Mice, Inbred C57BL , Mice, Knockout , Up-Regulation/drug effects , Up-Regulation/genetics , beta Catenin/metabolism
5.
Dis Model Mech ; 8(9): 1093-103, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26183215

ABSTRACT

Hypoxia-inducible factor 1α (HIF1α) is a transcription factor that regulates the adaptation of cells to hypoxic microenvironments, for example inside solid tumours. Stabilisation of HIF1α can also occur in normoxic conditions in inflamed tissue or as a result of inactivating mutations in negative regulators of HIF1α. Aberrant overexpression of HIF1α in many different cancers has led to intensive efforts to develop HIF1α-targeted therapies. However, the role of HIF1α is still poorly understood in chronic inflammation that predisposes the colon to carcinogenesis. We have previously reported that the transcription of HIF1α is upregulated and that the protein is stabilised in inflammatory lesions that are caused by the non-steroidal anti-inflammatory drug (NSAID) sulindac in the mouse proximal colon. Here, we exploited this side effect of long-term sulindac administration to analyse the role of HIF1α in colon inflammation using mice with a Villin-Cre-induced deletion of Hif1α exon 2 in the intestinal epithelium (Hif1α(ΔIEC)). We also analysed the effect of sulindac sulfide on the aryl hydrocarbon receptor (AHR) pathway in vitro in colon cancer cells. Most sulindac-treated mice developed visible lesions, resembling the appearance of flat adenomas in the human colon, surrounded by macroscopically normal mucosa. Hif1α(ΔIEC) mice still developed lesions but they were smaller than in the Hif1α-floxed siblings (Hif1α(F/F)). Microscopically, Hif1α(ΔIEC) mice had significantly less severe colon inflammation than Hif1α(F/F) mice. Molecular analysis showed reduced MIF expression and increased E-cadherin mRNA expression in the colon of sulindac-treated Hif1α(ΔIEC) mice. However, immunohistochemistry analysis revealed a defect of E-cadherin protein expression in sulindac-treated Hif1α(ΔIEC) mice. Sulindac sulfide treatment in vitro upregulated Hif1α, c-JUN and IL8 expression through the AHR pathway. Taken together, HIF1α expression augments inflammation in the proximal colon of sulindac-treated mice, and AHR activation by sulindac might lead to the reduction of E-cadherin protein levels through the mitogen-activated protein kinase (MAPK) pathway.


Subject(s)
Colonic Neoplasms/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Inflammation , Animals , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cadherins/metabolism , Cell Line, Tumor , Colonic Neoplasms/pathology , Disease Models, Animal , Exons , Female , Gene Deletion , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/deficiency , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Immunohistochemistry , Interleukin-8/metabolism , Intestinal Mucosa/pathology , MAP Kinase Signaling System , Male , Mice , Oncogene Protein p65(gag-jun)/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Sulindac/therapeutic use , Up-Regulation
6.
Biochem Biophys Res Commun ; 413(2): 336-41, 2011 Sep 23.
Article in English | MEDLINE | ID: mdl-21893047

ABSTRACT

We describe a coculture model of a human intestinal epithelial cell line and human peripheral blood monocytes in which monocytes differentiate into cells with features of resident intestinal macrophages. Caco-2 cells are grown on the lower surface of a semipermeable filter with pore size of 3 µm (Transwells) until they differentiate into enterocytes. Peripheral-blood monocytes are added and the co-culture incubated for two days. Monocytes migrate through the pores of the membrane, come into direct contact with the basolateral surfaces of the epithelial cell monolayer, and develop characteristics of resident intestinal macrophages including downregulation of CD14 expression and reduced pro-inflammatory cytokine responses (IL-8, TNF and IL-1ß) to bacterial products. The apical application of lipopolysaccharide (LPS) and muramyl dipeptide (MDP) resulted in an increased number of integrated monocytes, but abrogated the downregulation of CD14 expression and the diminished cytokine responses. MDP also reduced tight-junctional integrity, whilst LPS had no effect. These data indicate that LPS and MDP have significant pathophysiological effects on enterocyte-monocyte interactions, and confirm other studies that demonstrate that enterocytes and their products influence monocyte differentiation. This model may be useful in providing insights into the interaction between monocytes, epithelial cells and intestinal bacteria in health and disease.


Subject(s)
Acetylmuramyl-Alanyl-Isoglutamine/pharmacology , Enterocytes/drug effects , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Bacteria , Caco-2 Cells , Cell Nucleus/drug effects , Cell Nucleus/ultrastructure , Coculture Techniques , Cytokines/metabolism , Down-Regulation , Enterocytes/physiology , Enterocytes/ultrastructure , Humans , Lipopolysaccharide Receptors/metabolism , Macrophages/physiology , Macrophages/ultrastructure , Monocytes/drug effects , Monocytes/physiology , Tight Junctions
7.
Exp Eye Res ; 92(5): 361-76, 2011 May.
Article in English | MEDLINE | ID: mdl-21354137

ABSTRACT

Human fetal eyes 8-40 weeks gestation (WG) were examined using markers to hematopoietic stem cells (HSC), vascular precursor cells (VPC), monocytes/macrophages and endothelial cells (EC). Electron microscopy and bromo-deoxyuridene labeling were undertaken to confirm the existence of solid vascular cords and to demonstrate vasculogenesis and angiogenesis in developing choroidal tissue. Our results demonstrated that the earliest incipient choroid consisted of vimentin(+) mesenchymal precursor cells which downregulated vimentin expression with maturation. Our observations lead us to conclude that these vimentin(-)/CD34(+)/CD44(+)/CD133(+) HSCs then differentiated into three distinct lineages: single isolated CD34(-)/CD39(+) VPCs that formed solid vascular cords which lumenized and became lined with CD34(+) vascular ECs; CD34(--+)/CD14(+)/CD68(+) monocytes that differentiated into tissue macrophages; and CD133(+)/CD34(--+)/α-smooth muscle actin(+) mural precursor cells that matured into smooth muscle cells and pericytes. Blood vessel formation occurred throughout the whole choroid simultaneously, indicative of in situ differentiation. Vasculogenesis, as evidenced by lumenization of solid vascular cords, was responsible for the formation of the entire choroidal area with angiogenesis, in all three layers of the choroid, only adding to vascular density. These results suggest that formation of the human choroid involves three processes: HSC differentiation, vasculogenesis and angiogenesis. Since vasculogenesis takes place independently of VEGF(165), further insights regarding the molecular mechanisms of vasculogenesis are required to better inform future treatments of choroidal neovascularization.


Subject(s)
Cell Differentiation/physiology , Choroid/blood supply , Choroid/embryology , Endothelium, Vascular/cytology , Hematopoietic Stem Cells/cytology , Neovascularization, Physiologic/physiology , Actins/metabolism , Antigens, CD/metabolism , Biomarkers/metabolism , Capillaries/cytology , Capillaries/metabolism , Cell Lineage , Endothelium, Vascular/metabolism , Gestational Age , Hematopoietic Stem Cells/metabolism , Humans , Immunohistochemistry , Ki-67 Antigen/metabolism , Macrophages/cytology , Mesenchymal Stem Cells/cytology , Microscopy, Confocal , Microscopy, Electron , Vimentin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...