Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Biochem Biophys Res Commun ; 629: 34-39, 2022 11 12.
Article in English | MEDLINE | ID: mdl-36099782

ABSTRACT

Tongue epithelium is one of the most proliferative and regenerative epithelia in our body. However, tongue stem cell research is hampered partly by the lack of optimal animal models to study tongue injury, repair, and regeneration. Here, we establish a novel chemically induced tongue injury-recovery mouse model. Focal application of sodium hydroxide for a limited time led to the denudation of suprabasal layers, leaving the basal layer. Time course study revealed that tongue epithelial cells robustly proliferate over one week after the tongue injury. Importantly, we demonstrated that our novel mouse model could be employed in the lineage tracing of the tongue stem cells under the injury and repair process and further showed that tongue stem cells proliferate faster and generate larger clones in the injury condition than in the steady state condition. Our data indicate the development of a novel chemically induced tongue injury-recovery mouse model for tongue stem cell research, which will significantly facilitate the preclinical study for the pathogenesis and treatment of caustic ingestion.


Subject(s)
Caustics , Animals , Epithelial Cells , Epithelium , Mice , Sodium Hydroxide , Tongue
2.
Biomaterials ; 289: 121765, 2022 Oct.
Article in English | MEDLINE | ID: mdl-36067566

ABSTRACT

Extracellular vesicles (EVs) mediate cell-cell crosstalk by carrying bioactive molecules derived from cells. Recently, immune cell-derived EVs have been reported to regulate key biological functions such as tumor progression. CD4+ T cells orchestrate overall immunity; however, the biological role of their EVs is unclear. This study reveals that EVs derived from CD4+ T cells increase the antitumor response of CD8+ T cells by enhancing their proliferation and activity without affecting regulatory T cells (Tregs). Moreover, EVs derived from interleukin-2 (IL2)-stimulated CD4+ T cells induce a more enhanced antitumor response of CD8+ T cells compared with that of IL2-unstimulated CD4+ T cell-derived EVs. Mechanistically, miR-25-3p, miR-155-5p, miR-215-5p, and miR-375 within CD4+ T cell-derived EVs are responsible for the induction of CD8+ T cell-mediated antitumor responses. In a melanoma mouse model, the EVs potently suppress tumor growth through CD8+ T cell activation. This study demonstrates that the EVs, in addition to IL2, are important mediators between CD4+ and CD8+ T cells. Furthermore, unlike IL2, clinically used as an antitumor agent, CD4+ T cell-derived EVs stimulate CD8+ T cells without activating Tregs. Therefore, CD4+ T cell-derived EVs may provide a novel direction for cancer immunotherapy by inducing a CD8+ T cell-mediated antitumor response.


Subject(s)
Extracellular Vesicles , MicroRNAs , Animals , CD4-Positive T-Lymphocytes , CD8-Positive T-Lymphocytes , Interleukin-2 , Mice , T-Lymphocytes, Regulatory
3.
Cytokine ; 143: 155542, 2021 07.
Article in English | MEDLINE | ID: mdl-33926775

ABSTRACT

Interferon-γ (IFNG) is one of the key cytokines that regulates both innate and adaptive immune responses in the body. However, the role of IFNG in the regulation of vascularization, especially in the context of Vascular endothelial growth factor A (VEGFa)-induced angiogenesis is not clarified. Here, we report that IFNG shows potent anti-angiogenic potential against VEGFa-induced angiogenesis. IFNG significantly inhibited proliferation, migration, and tube formation of Human umbilical vein endothelial cells (HUVECs) both under basal and VEGFa-treated conditions. Intriguingly, Knockdown (KD) of STAT1 abolished the inhibitory effect of IFNG on VEGFa-induced angiogenic processes in HUVECs. Furthermore, IFNG exhibited potent anti-angiogenic efficacy in the mouse model of oxygen-induced retinopathy (OIR), an in vivo model for hypoxia-induced retinal neovascularization, without induction of functional side effects. Taken together, these results show that IFNG plays a crucial role in the regulation of VEGFa-dependent angiogenesis, suggesting its potential therapeutic applicability in neovascular diseases.


Subject(s)
Interferon-gamma/therapeutic use , Ischemia/complications , Retinal Neovascularization/complications , Retinal Neovascularization/drug therapy , Animals , Cell Movement/drug effects , Cell Proliferation/drug effects , Disease Models, Animal , Down-Regulation/drug effects , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Hypoxia/complications , Interferon-gamma/administration & dosage , Interferon-gamma/pharmacology , Intravitreal Injections , Mice , Neovascularization, Physiologic/drug effects , Retina/drug effects , Retina/pathology , Retina/physiopathology , Retinal Neovascularization/physiopathology , STAT1 Transcription Factor/metabolism , Signal Transduction/drug effects
4.
Biochem Biophys Res Commun ; 558: 29-35, 2021 06 18.
Article in English | MEDLINE | ID: mdl-33895548

ABSTRACT

Estrogen therapy is used to treat patients with post-menopausal symptoms, such as hot flashes and dyspareunia. Estrogen therapy also decreases the risk of fractures from osteoporosis in post-menopausal women. However, estrogen increases the risk of venous thromboembolic events, such as pulmonary embolism, but the pathways through which estrogen increase the risk of thromboembolism is unknown. Here, we show that estrogen elicits endothelial exocytosis, the key step in vascular thrombosis and inflammation. Exogenous 17ß-estradiol (E2) stimulated endothelial exocytosis of Weibel-Palade bodies (WPBs), releasing von Willebrand factor (vWF) and interleukin-8 (IL-8). Conversely, the estrogen antagonist ICI-182,780 interfered with E2-induced endothelial exocytosis. The ERα agonist propyl pyrazole triol (PPT) but not the ERß agonist diarylpropionitrile (DPN) induced vWF release, while ERα silencing counteracted vWF release by E2, suggesting that ERα mediates this effect. Exocytosis triggered by E2 occurred rapidly within 15 min and was not inhibited by either actinomycin D or cycloheximide. On the contrary, it was inhibited by the pre-treatment of U0126 or SB203580, an ERK or a p38 inhibitor, respectively, suggesting that E2-induced endothelial exocytosis is non-genomically mediated by the MAP kinase pathway. Finally, E2 treatment enhanced platelet adhesion to endothelial cells ex vivo, which was interfered with the pre-treatment of ICI-182,780 or U0126. Taken together, our data show that estrogen activates endothelial exocytosis non-genomically through the ERα-MAP kinase pathway. Our data suggest that adverse cardiovascular effects such as vascular inflammation and thrombosis should be considered in patients before menopausal hormone treatment.


Subject(s)
Endothelial Cells/drug effects , Estradiol/adverse effects , Exocytosis/drug effects , Endothelial Cells/pathology , Endothelial Cells/physiology , Estradiol/pharmacology , Estrogen Receptor alpha/metabolism , Estrogen Replacement Therapy/adverse effects , Exocytosis/physiology , Female , Human Umbilical Vein Endothelial Cells , Humans , In Vitro Techniques , MAP Kinase Signaling System/drug effects , Platelet Adhesiveness/drug effects , Platelet Adhesiveness/physiology , Postmenopause/drug effects , Postmenopause/physiology , Risk Factors , Thromboembolism/etiology , Weibel-Palade Bodies/drug effects , Weibel-Palade Bodies/pathology , Weibel-Palade Bodies/physiology
5.
Genome Biol ; 21(1): 107, 2020 05 07.
Article in English | MEDLINE | ID: mdl-32381040

ABSTRACT

BACKGROUND: Tumors comprise a complex microenvironment of interacting malignant and stromal cell types. Much of our understanding of the tumor microenvironment comes from in vitro studies isolating the interactions between malignant cells and a single stromal cell type, often along a single pathway. RESULT: To develop a deeper understanding of the interactions between cells within human lung tumors, we perform RNA-seq profiling of flow-sorted malignant cells, endothelial cells, immune cells, fibroblasts, and bulk cells from freshly resected human primary non-small-cell lung tumors. We map the cell-specific differential expression of prognostically associated secreted factors and cell surface genes, and computationally reconstruct cross-talk between these cell types to generate a novel resource called the Lung Tumor Microenvironment Interactome (LTMI). Using this resource, we identify and validate a prognostically unfavorable influence of Gremlin-1 production by fibroblasts on proliferation of malignant lung adenocarcinoma cells. We also find a prognostically favorable association between infiltration of mast cells and less aggressive tumor cell behavior. CONCLUSION: These results illustrate the utility of the LTMI as a resource for generating hypotheses concerning tumor-microenvironment interactions that may have prognostic and therapeutic relevance.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , Cell Communication , Lung Neoplasms/metabolism , Receptor Cross-Talk , Tumor Microenvironment , Adenocarcinoma/metabolism , Cell Line, Tumor , Fibroblasts/metabolism , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Primary Cell Culture
6.
Clin Cancer Res ; 26(1): 274-281, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31548347

ABSTRACT

PURPOSE: Activation of NFE2L2 has been linked to chemoresistance in cell line models. Recently, somatic mutations that activate NFE2L2, including mutations in NFE2L2, KEAP1, or CUL3, have been found to be associated with poor outcomes in patients with non-small cell lung cancer (NSCLC). However, the impact of these mutations on chemoresistance remains incompletely explored. EXPERIMENTAL DESIGN: We investigated the effect of Keap1 deletion on chemoresistance in cell lines from Trp53-based mouse models of lung squamous cell carcinoma (LSCC) and lung adenocarcinoma (LUAD). Separately, we identified 51 patients with stage IV NSCLC with KEAP1, NFE2L2, or CUL3 mutations and a matched cohort of 52 wild-type patients. Time to treatment failure after first-line platinum doublet chemotherapy and overall survival was compared between the two groups. RESULTS: Deletion of Keap1 in Trp53-null murine LUAD and LSCC resulted in increased clonogenic survival upon treatment with diverse cytotoxic chemotherapies. In patients with NSCLC, median time to treatment failure (TTF) after first-line chemotherapy for the KEAP1/NFE2L2/CUL3-mutant cohort was 2.8 months compared with 8.3 months in the control group (P < 0.0001). Median overall survival (OS) was 11.2 months in the KEAP1/NFE2L2/CUL3-mutant group and 36.8 months in the control group (P = 0.006). CONCLUSIONS: Keap1 deletion confers chemoresistance in murine lung cancer cells. Patients with metastatic NSCLC with mutations in KEAP1, NFE2L2, or CUL3 have shorter TTF and OS after first-line platinum doublet chemotherapy compared with matched controls. Novel approaches for improving outcomes in this subset of patients with NSCLC are therefore needed.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Non-Small-Cell Lung/pathology , Drug Resistance, Neoplasm/genetics , Kelch-Like ECH-Associated Protein 1/genetics , Lung Neoplasms/pathology , Mutation , NF-E2-Related Factor 2/genetics , Aged , Animals , Carboplatin/administration & dosage , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Cisplatin/administration & dosage , Etoposide/administration & dosage , Female , Humans , Kelch-Like ECH-Associated Protein 1/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-E2-Related Factor 2/metabolism , Neoplasm Staging , Paclitaxel/administration & dosage , Prognosis , Spheroids, Cellular , Survival Rate
7.
Cytokine ; 126: 154863, 2020 02.
Article in English | MEDLINE | ID: mdl-31629112

ABSTRACT

Adiponectin (Ad) is a representative adipocytokine that regulates energy homeostasis including glucose transport and lipid oxidation through activation of AMP-activated protein kinase (AMPK) pathways. Plasma levels of Ad are reduced in obesity, which contributes to type 2 diabetes. Therefore, agents that activate the Ad signaling pathway could ameliorate metabolic diseases such as type 2 diabetes. Here, we report the identification of a high-affinitive agonist antibody against Ad receptors. The antibody was selected by using phage display of human combinatorial antibody libraries. The selected antibody induced phosphorylation of the acetyl-CoA carboxylase (ACC) and AMPK in skeletal muscle cells and stimulated glucose uptake and fatty-acid oxidation (FAO) in myotubes. In addition, the antibody significantly lowered blood glucose levels during a glucose challenge in normal mice as well as basal blood glucose levels in a type 2 diabetic mouse model. Taken together, these results suggest that the agonist antibody could be a promising therapeutic agent for the treatment of metabolic syndrome such as type 2 diabetes.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Fatty Acids/metabolism , Glucose/metabolism , Receptors, Adiponectin/agonists , Receptors, Adiponectin/immunology , Acetyl-CoA Carboxylase/metabolism , Adiponectin/metabolism , Animals , Blood Glucose/metabolism , Cell Line , Diabetes Mellitus, Type 2/metabolism , Gene Knockdown Techniques , Glucose/pharmacology , Humans , Lipid Metabolism , Male , Mice , Mice, Inbred C57BL , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/metabolism , NF-kappa B/metabolism , Oxidation-Reduction , Phosphorylation , RNA, Small Interfering , Receptors, Adiponectin/genetics , Signal Transduction/drug effects , Signal Transduction/genetics
8.
Nat Commun ; 9(1): 5110, 2018 11 30.
Article in English | MEDLINE | ID: mdl-30504895

ABSTRACT

Non-small cell lung carcinoma (NSCLC) is leading cause of cancer-related deaths in the world. The Tumor Suppressor Candidate 3 (TUSC3) at chromosome 8p22 known to be frequently deleted in cancer is often found to be deleted in advanced stage of solid tumors. However, the role of TUSC3 still remains controversial in lung cancer and context-dependent in several cancers. Here we propose that miR-224/-520c-dependent TUSC3 deficiency enhances the metastatic potential of NSCLC through the alteration of three unfolded protein response pathways and HRD1-dependent ERAD. ATF6α-dependent UPR is enhanced whereas the affinity of HRD1 to its substrates, PERK, IRE1α and p53 is weakened. Consequently, the alteration of UPRs and the suppressed p53-NM23H1/2 pathway by TUSC3 deficiency is ultimately responsible for enhancing metastatic potential of lung cancer. These findings provide mechanistic insight of unrecognized roles of TUSC3 in cancer progression and the oncogenic role of HRD1-dependent ERAD in cancer metastasis.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Endoplasmic Reticulum-Associated Degradation/physiology , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Membrane Proteins/metabolism , MicroRNAs/metabolism , Tumor Suppressor Proteins/metabolism , Activating Transcription Factor 6/genetics , Activating Transcription Factor 6/metabolism , Animals , Carcinoma, Non-Small-Cell Lung/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Cell Proliferation/physiology , Endoplasmic Reticulum-Associated Degradation/genetics , Endoribonucleases/genetics , Endoribonucleases/metabolism , Gene Expression Regulation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic/physiology , Humans , In Situ Hybridization , Lung Neoplasms/genetics , Membrane Proteins/genetics , Mice , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/genetics , Signal Transduction/physiology , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Unfolded Protein Response/genetics , Unfolded Protein Response/physiology , Xenograft Model Antitumor Assays
9.
Nat Commun ; 9(1): 368, 2018 01 25.
Article in English | MEDLINE | ID: mdl-29371594

ABSTRACT

During development, the formation of a mature, well-functioning heart requires transformation of the ventricular wall from a loose trabecular network into a dense compact myocardium at mid-gestation. Failure to compact is associated in humans with congenital diseases such as left ventricular non-compaction (LVNC). The mechanisms regulating myocardial compaction are however still poorly understood. Here, we show that deletion of the Ino80 chromatin remodeler in vascular endothelial cells prevents ventricular compaction in the developing mouse heart. This correlates with defective coronary vascularization, and specific deletion of Ino80 in the two major coronary progenitor tissues-sinus venosus and endocardium-causes intermediate phenotypes. In vitro, endothelial cells promote myocardial expansion independently of blood flow in an Ino80-dependent manner. Ino80 deletion increases the expression of E2F-activated genes and endothelial cell S-phase occupancy. Thus, Ino80 is essential for coronary angiogenesis and allows coronary vessels to support proper compaction of the heart wall.


Subject(s)
Adenosine Triphosphatases/metabolism , Endothelium, Vascular/metabolism , Heart Defects, Congenital/metabolism , Neovascularization, Pathologic/metabolism , ATPases Associated with Diverse Cellular Activities , Adenosine Triphosphatases/genetics , Animals , Coronary Vessels/metabolism , DNA Helicases/genetics , DNA Helicases/metabolism , DNA-Binding Proteins , Endocardium/metabolism , Endocardium/pathology , Endothelial Cells/enzymology , Endothelial Cells/metabolism , Endothelium, Vascular/pathology , Heart Defects, Congenital/genetics , Heart Ventricles/metabolism , Heart Ventricles/pathology , Humans , Mice, Knockout , Mice, Transgenic , Myocardium/metabolism , Myocardium/pathology , Neovascularization, Pathologic/genetics
10.
Cancer Discov ; 7(1): 86-101, 2017 01.
Article in English | MEDLINE | ID: mdl-27663899

ABSTRACT

Lung squamous cell carcinoma (LSCC) pathogenesis remains incompletely understood, and biomarkers predicting treatment response remain lacking. Here, we describe novel murine LSCC models driven by loss of Trp53 and Keap1, both of which are frequently mutated in human LSCCs. Homozygous inactivation of Keap1 or Trp53 promoted airway basal stem cell (ABSC) self-renewal, suggesting that mutations in these genes lead to expansion of mutant stem cell clones. Deletion of Trp53 and Keap1 in ABSCs, but not more differentiated tracheal cells, produced tumors recapitulating histologic and molecular features of human LSCCs, indicating that they represent the likely cell of origin in this model. Deletion of Keap1 promoted tumor aggressiveness, metastasis, and resistance to oxidative stress and radiotherapy (RT). KEAP1/NRF2 mutation status predicted risk of local recurrence after RT in patients with non-small lung cancer (NSCLC) and could be noninvasively identified in circulating tumor DNA. Thus, KEAP1/NRF2 mutations could serve as predictive biomarkers for personalization of therapeutic strategies for NSCLCs. SIGNIFICANCE: We developed an LSCC mouse model involving Trp53 and Keap1, which are frequently mutated in human LSCCs. In this model, ABSCs are the cell of origin of these tumors. KEAP1/NRF2 mutations increase radioresistance and predict local tumor recurrence in radiotherapy patients. Our findings are of potential clinical relevance and could lead to personalized treatment strategies for tumors with KEAP1/NRF2 mutations. Cancer Discov; 7(1); 86-101. ©2016 AACR.This article is highlighted in the In This Issue feature, p. 1.


Subject(s)
Carcinoma, Squamous Cell/genetics , Kelch-Like ECH-Associated Protein 1/genetics , Lung Neoplasms/genetics , NF-E2-Related Factor 2/genetics , Radiation Tolerance , Trachea/pathology , Tumor Suppressor Protein p53/genetics , Adult , Aged , Aged, 80 and over , Animals , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/radiotherapy , Cell Line, Tumor , Cell Self Renewal , Disease Models, Animal , Female , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/pathology , Lung Neoplasms/radiotherapy , Male , Mice , Middle Aged , Mutation , Neoplasm Invasiveness , Stem Cells/cytology , Stem Cells/pathology , Trachea/cytology , Tumor Cells, Cultured
11.
Oncotarget ; 7(27): 41798-41810, 2016 Jul 05.
Article in English | MEDLINE | ID: mdl-27260001

ABSTRACT

Hexokinase 2 (HK2) is a rate-determining enzyme in aerobic glycolysis, a process upregulated in tumor cells. HK2 expression is controlled by various transcription factors and epigenetic alterations and is heterogeneous in hepatocellular carcinomas (HCCs), though the cause of this heterogeneity is not known. DNA methylation in the HK2 promoter CpG island (HK2-CGI) and its surrounding regions (shore and shelf) has not previously been evaluated, but may provide clues about the regulation of HK2 expression. Here, we compared HK2 promoter methylation in HCCs and adjacent non-cancerous liver tissues using a HumanMethylation450 BeadChip array. We found that, while the HK2-CGI N-shore was hypomethylated, thereby enhancing HK2 expression, the HK2-CGI was itself hypermethylated in some HCCs. This hypermethylation suppressed HK2 expression by inhibiting interactions between HIF-1α and a hypoxia response element (HRE) located at -234/-230. HCCs that were HK2negative and had distinct promoter CGI methylation were denoted as having a HK2-CGI methylation phenotype (HK2-CIMP), which was associated with poor clinical outcome. These findings indicate that HK2-CGI N-shore hypomethylation and HK2-CGI hypermethylation affect HK2 expression by influencing the interaction between HIF 1α and HRE. HK2-CGI hypermethylation induces HK2-CIMP and could represent a prognostic biomarker for HCC.


Subject(s)
Carcinoma, Hepatocellular/genetics , CpG Islands/genetics , DNA Methylation , Hexokinase/genetics , Liver Neoplasms/genetics , Promoter Regions, Genetic , Biomarkers, Tumor/genetics , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Disease Progression , Gene Expression Regulation, Neoplastic , Hexokinase/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Liver/metabolism , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Prognosis
12.
Gut ; 65(7): 1077-86, 2016 07.
Article in English | MEDLINE | ID: mdl-25897018

ABSTRACT

OBJECTIVE: Human oesophageal stem cell research is hampered by the lack of an optimal assay system to study self-renewal and differentiation. We aimed to identify and characterise human and mouse oesophageal stem/progenitor cells by establishing 3-dimensional organotypic sphere culture systems for both species. DESIGN: Primary oesophageal epithelial cells were freshly isolated and fluorescence-activated cell sorting (FACS)-sorted from human and mouse oesophagus and 3-dimensional organotypic sphere culture systems were developed. The self-renewing potential and differentiation status of novel subpopulations were assessed by sphere-forming ability, cell cycle analysis, immunostaining, qPCR and RNA-Seq. RESULTS: Primary human and mouse oesophageal epithelial cells clonally formed esophagospheres consisting of stratified squamous epithelium. Sphere-forming cells could self-renew and form esophagospheres for over 43 passages in vitro and generated stratified squamous epithelium when transplanted under the kidney capsule of immunodeficient mice. Sphere-forming cells were 10-15-fold enriched among human CD49f(hi)CD24(low) cells and murine CD49f(+)CD24(low)CD71(low) cells compared with the most differentiated cells. Genetic elimination of p63 in mouse and human oesophageal cells dramatically decreased esophagosphere formation and basal gene expression while increasing suprabasal gene expression. CONCLUSIONS: We developed clonogenic and organotypic culture systems for the quantitative analyses of human and mouse oesophageal stem/progenitor cells and identified novel cell surface marker combinations that enrich for these cells. Using this system, we demonstrate that elimination of p63 inhibits self-renewal of human oesophageal stem/progenitor cells. We anticipate that these esophagosphere culture systems will facilitate studies of oesophageal stem cell biology and may prove useful for ex vivo expansion of human oesophageal stem cells.


Subject(s)
Cell Self Renewal/genetics , Epithelial Cells/physiology , Epithelium/growth & development , Esophagus/cytology , Spheroids, Cellular/cytology , Stem Cells/physiology , Animals , Antigens, CD/analysis , Antigens, CD/genetics , CD24 Antigen/analysis , CD24 Antigen/genetics , Cell Differentiation/genetics , Epithelial Cells/cytology , Gene Expression , Humans , Integrin alpha6/analysis , Integrin alpha6/genetics , Mice , Mice, Knockout , Phosphoproteins/genetics , Primary Cell Culture/methods , Receptors, Transferrin/analysis , Receptors, Transferrin/genetics , Spheroids, Cellular/transplantation , Stem Cells/chemistry , Stem Cells/cytology , Trans-Activators/genetics , Transcription Factors/genetics , Tumor Suppressor Proteins/genetics
13.
Mol Cancer Ther ; 14(8): 1951-61, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25934710

ABSTRACT

The anticancer actions of vitamin D and its hormonally active form, calcitriol, have been extensively documented in clinical and preclinical studies. However, the mechanisms underlying these actions have not been completely elucidated. Here, we examined the effect of dietary vitamin D and calcitriol on mouse breast tumor-initiating cells (TICs, also known as cancer stem cells). We focused on MMTV-Wnt1 mammary tumors, for which markers for isolating TICs have previously been validated. We confirmed that these tumors expressed functional vitamin D receptors and estrogen receptors (ER) and exhibited calcitriol-induced molecular responses including ER downregulation. Following orthotopic implantation of MMTV-Wnt1 mammary tumor cells into mice, calcitriol injections or a vitamin D-supplemented diet caused a striking delay in tumor appearance and growth, whereas a vitamin D-deficient diet accelerated tumor appearance and growth. Calcitriol inhibited TIC tumor spheroid formation in a dose-dependent manner in primary cultures and inhibited TIC self-renewal in secondary passages. A combination of calcitriol and ionizing radiation inhibited spheroid formation more than either treatment alone. Further, calcitriol significantly decreased TIC frequency as evaluated by in vivo limiting dilution analyses. Calcitriol inhibition of TIC spheroid formation could be overcome by the overexpression of ß-catenin, suggesting that the inhibition of Wnt/ß-catenin pathway is an important mechanism mediating the TIC inhibitory activity of calcitriol in this tumor model. Our findings indicate that vitamin D compounds target breast TICs reducing tumor-initiating activity. Our data also suggest that combining vitamin D compounds with standard therapies may enhance anticancer activity and improve therapeutic outcomes.


Subject(s)
Calcitriol/pharmacology , Neoplastic Stem Cells/drug effects , Vitamin D/pharmacology , Animals , Body Weight , Calcium/blood , Cell Line, Tumor , Estrogens/metabolism , Female , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/mortality , Mammary Neoplasms, Experimental/pathology , Mice , Neoplastic Stem Cells/metabolism , Receptors, Calcitriol/metabolism , Receptors, Estrogen/metabolism , Tumor Burden , Vitamin D/metabolism , Wnt Signaling Pathway/drug effects
14.
J Clin Invest ; 124(2): 553-63, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24435044

ABSTRACT

Erythropoietin (EPO) is a hormone that induces red blood cell production. In its recombinant form, EPO is the one of most prescribed drugs to treat anemia, including that arising in cancer patients. In randomized trials, EPO administration to cancer patients has been associated with decreased survival. Here, we investigated the impact of EPO modulation on tumorigenesis. Using genetically engineered mouse models of breast cancer, we found that EPO promoted tumorigenesis by activating JAK/STAT signaling in breast tumor-initiating cells (TICs) and promoted TIC self renewal. We determined that EPO was induced by hypoxia in breast cancer cell lines, but not in human mammary epithelial cells. Additionally, we demonstrated that high levels of endogenous EPO gene expression correlated with shortened relapse-free survival and that pharmacologic JAK2 inhibition was synergistic with chemotherapy for tumor growth inhibition in vivo. These data define an active role for endogenous EPO in breast cancer progression and breast TIC self-renewal and reveal a potential application of EPO pathway inhibition in breast cancer therapy.


Subject(s)
Breast Neoplasms/metabolism , Erythropoietin/metabolism , Neoplastic Stem Cells/drug effects , Animals , Breast Neoplasms/therapy , Carcinogenesis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Transformation, Neoplastic/drug effects , Disease Progression , Disease-Free Survival , Endothelial Cells/metabolism , Female , Gene Expression Regulation , Humans , Hypoxia , Mammary Neoplasms, Experimental/drug therapy , Mice , Neoplasm Transplantation , Neoplastic Stem Cells/metabolism , Recurrence , Signal Transduction , Tetrazolium Salts , Thiazoles , Time Factors
15.
J Leukoc Biol ; 95(4): 651-9, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24374966

ABSTRACT

The MAPK pathway mediates TLR signaling during innate immune responses. We discovered previously that MKP-1 is acetylated, enhancing its interaction with its MAPK substrates and deactivating TLR signaling. As HDACs modulate inflammation by deacetylating histone and nonhistone proteins, we hypothesized that HDACs may regulate LPS-induced inflammation by deacetylating MKP-1. We found that mouse macrophages expressed a subset of HDAC isoforms (HDAC1, HDAC2, and HDAC3), which all interacted with MKP-1. Genetic silencing or pharmacologic inhibition of HDAC1, -2, and -3 increased MKP-1 acetylation in cells. Furthermore, knockdown or pharmacologic inhibition of HDAC1, -2, and -3 decreased LPS-induced phosphorylation of the MAPK member p38. Also, pharmacologic inhibition of HDAC did not decrease MAPK signaling in MKP-1 null cells. Finally, inhibition of HDAC1, -2, and -3 decreased LPS-induced expression of TNF-α, IL-1ß, iNOS (NOS2), and nitrite synthesis. Taken together, our results show that HDAC1, -2, and -3 deacetylate MKP-1 and that this post-translational modification increases MAPK signaling and innate immune signaling. Thus, HDAC1, -2, and -3 isoforms are potential therapeutic targets in inflammatory diseases.


Subject(s)
Dual Specificity Phosphatase 1/metabolism , Histone Deacetylases/physiology , Immunity, Innate , Acetylation , Amino Acid Sequence , Animals , Cells, Cultured , Cytokines/biosynthesis , Inflammation/etiology , Isoenzymes/physiology , Lipopolysaccharides/pharmacology , MAP Kinase Signaling System , Macrophages/enzymology , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Phosphorylation , p38 Mitogen-Activated Protein Kinases/metabolism
16.
Proc Natl Acad Sci U S A ; 106(10): 3782-7, 2009 Mar 10.
Article in English | MEDLINE | ID: mdl-19223584

ABSTRACT

Although elevated levels of aldosterone are associated with vascular inflammation, the proinflammatory pathways of aldosterone are not completely defined. We now show that aldosterone triggers endothelial cell exocytosis, the first step in leukocyte trafficking. Exogenous aldosterone stimulates endothelial exocytosis of Weibel-Palade bodies, externalizing P-selectin and releasing von Willebrand factor. Spironolactone, a nonselective mineralocorticoid receptor (MR) blocker, antagonizes aldosterone-induced endothelial exocytosis. Knockdown of the MR also decreases exocytosis, suggesting that the MR mediates exocytosis. Aldosterone triggers exocytosis within minutes, and this effect is not inhibited by actinomycin D, suggesting a nongenomic effect of aldosterone. Aldosterone treatment of endothelial cells increases leukocyte adherence to endothelial cells in culture. Taken together, our data suggest that aldosterone activates vascular inflammation in part through nongenomic, MR-mediated pathways. Aldosterone antagonism may decrease vascular inflammation and cardiac fibrosis in part by blocking endothelial exocytosis.


Subject(s)
Aldosterone/pharmacology , Endothelial Cells/cytology , Endothelial Cells/drug effects , Exocytosis/drug effects , Calcium Signaling/drug effects , Cell Communication/drug effects , Gene Expression Regulation/drug effects , HeLa Cells , Humans , Leukocytes/cytology , Leukocytes/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Mineralocorticoid/genetics , Receptors, Mineralocorticoid/metabolism , Weibel-Palade Bodies/drug effects , Weibel-Palade Bodies/metabolism
17.
J Biochem Mol Biol ; 39(1): 84-90, 2006 Jan 31.
Article in English | MEDLINE | ID: mdl-16466642

ABSTRACT

During our search for macrophage stimulating compounds from medicinal plants, we isolated biopolymers from Acanthopanax sessiliflorus. Isolated fraction AS-5 showed maximum potential, and stimulated lysosonal enzymatic activity by 230% at 300 microg/ml. The nitric oxide (NO) producing ability of AS-5 100 microg/ml was 58 microM when treated with interferon-gamma and lipopolysaccharide 20 micro/ml. The lymphocyte proliferating effects of isolated biopolymer fractions were also investigated. Highest lymphoproliferative activity (a 2.8-fold enhancement compared to salines treated group was exhibited by AS-3 at 200 micro/ml followed by AS-5 and AS-6. The AS-3 fraction stimulated only T-lymphocytes and had little or no effect on B-lymphocyte proliferation. Partially methylated alditol acetates were prepared to elucidate the glycosyl linkage-compositions of the AS-3 and AS-5 biopolymers, and were analyzed by GC-MS. The AS-3 and AS-5 biopolymer fractions were found to contain 2,3,4-tri-O-methyl-D-glucitol, 2,3,4-tri-O-methyl-D-galacitol 3,4,6-tri-O-methyl-galacitol, 2-O-methyl-arabinitol and 2,4,6-tri-O-methyl-D-glucitol, 2,3,6-tri-O-methyl-D-galacitol linkages, respectively.


Subject(s)
Biopolymers , Eleutherococcus/chemistry , Immunologic Factors , Plants, Medicinal/chemistry , Animals , Biopolymers/chemistry , Biopolymers/immunology , Cell Proliferation , Cells, Cultured , Immunologic Factors/chemistry , Immunologic Factors/immunology , Lymphocytes/cytology , Lymphocytes/immunology , Macrophages/cytology , Macrophages/enzymology , Macrophages/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Muramidase/metabolism , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/metabolism , Spleen/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...