Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Nat Commun ; 15(1): 5873, 2024 Jul 12.
Article in English | MEDLINE | ID: mdl-38997291

ABSTRACT

Low response rate, treatment relapse, and resistance remain key challenges for cancer treatment with immune checkpoint blockade (ICB). Here we report that loss of specific tumor suppressors (TS) induces an inflammatory response and promotes an immune suppressive tumor microenvironment. Importantly, low expression of these TSs is associated with a higher expression of immune checkpoint inhibitory mediators. Here we identify, by using in vivo CRISPR/Cas9 based loss-of-function screening, that NF1, TSC1, and TGF-ß RII as TSs regulating immune composition. Loss of each of these three TSs leads to alterations in chromatin accessibility and enhances IL6-JAK3-STAT3/6 inflammatory pathways. This results in an immune suppressive landscape, characterized by increased numbers of LAG3+ CD8 and CD4 T cells. ICB targeting LAG3 and PD-L1 simultaneously inhibits metastatic progression in preclinical triple negative breast cancer (TNBC) mouse models of NF1-, TSC1- or TGF-ß RII- deficient tumors. Our study thus reveals a role of TSs in regulating metastasis via non-cell-autonomous modulation of the immune compartment and provides proof-of-principle for ICB targeting LAG3 for patients with NF1-, TSC1- or TGF-ß RII-inactivated cancers.


Subject(s)
B7-H1 Antigen , Immune Checkpoint Inhibitors , Lymphocyte Activation Gene 3 Protein , Triple Negative Breast Neoplasms , Tuberous Sclerosis Complex 1 Protein , Tumor Microenvironment , Tumor Microenvironment/immunology , Animals , Mice , Female , Humans , Triple Negative Breast Neoplasms/immunology , Triple Negative Breast Neoplasms/pathology , Triple Negative Breast Neoplasms/genetics , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Tuberous Sclerosis Complex 1 Protein/genetics , Tuberous Sclerosis Complex 1 Protein/metabolism , B7-H1 Antigen/metabolism , B7-H1 Antigen/genetics , Neurofibromin 1/genetics , Neurofibromin 1/metabolism , Cell Line, Tumor , CD8-Positive T-Lymphocytes/immunology , Inflammation/immunology , CD4-Positive T-Lymphocytes/immunology , Gene Expression Regulation, Neoplastic , CRISPR-Cas Systems
2.
Neurospine ; 21(2): 443-454, 2024 06.
Article in English | MEDLINE | ID: mdl-38955522

ABSTRACT

OBJECTIVE: The study compared the morphometric changes of the cervical spinal cord using dynamic magnetic resonance imaging (MRI) in patients with cervical spondylotic myelopathy (CSM) and assessed the correlation with kinematic changes, cord cross-sectional area (CSA), and high signal intensity (SI) on T2-weighted imaging (T2WI). METHODS: Patients with CSM were evaluated through dynamic MRI for sagittal and axial CSA changes of the cervical cord, cerebrospinal fluid (CSF) reserve ratio, degree of cord impingement, cord compression rate, range of motion (ROM), and severity of SI on T2WI. The degree of cord impingement was evaluated using the Muhle grading system. Clinical outcomes were assessed using Japanese Orthopaedic Association scoring and Nurick grade. RESULTS: The study included 191 patients (113 males) with a mean age of 55.34 ± 12.09 years. The lowest sagittal CSF reserve ratio and cord occupation rate were observed during extension. Cord impingement and SI change were more prevalent in extension-positioned MRI. There was no difference between ROM on dynamic radiographs and dynamic MRI. Preoperative cervical ROM was greater in patients with intensely high SI change. CONCLUSION: Dynamic MRI is useful for evaluating neck movement. Patients with high SI had greater ROM before surgery but worse outcomes after. Neck extension exacerbated cervical stenosis and cord compression compared to flexion, and cervical spinal motion contributed to the severity of CSM. Cervical spinal motion should be carefully evaluated, particularly in hyperextension, to prevent worsening of CSM.

3.
Bone Joint Res ; 12(4): 245-255, 2023 Apr 03.
Article in English | MEDLINE | ID: mdl-37051826

ABSTRACT

To determine the major risk factors for unplanned reoperations (UROs) following corrective surgery for adult spinal deformity (ASD) and their interactions, using machine learning-based prediction algorithms and game theory. Patients who underwent surgery for ASD, with a minimum of two-year follow-up, were retrospectively reviewed. In total, 210 patients were included and randomly allocated into training (70% of the sample size) and test (the remaining 30%) sets to develop the machine learning algorithm. Risk factors were included in the analysis, along with clinical characteristics and parameters acquired through diagnostic radiology. Overall, 152 patients without and 58 with a history of surgical revision following surgery for ASD were observed; the mean age was 68.9 years (SD 8.7) and 66.9 years (SD 6.6), respectively. On implementing a random forest model, the classification of URO events resulted in a balanced accuracy of 86.8%. Among machine learning-extracted risk factors, URO, proximal junction failure (PJF), and postoperative distance from the posterosuperior corner of C7 and the vertical axis from the centroid of C2 (SVA) were significant upon Kaplan-Meier survival analysis. The major risk factors for URO following surgery for ASD, i.e. postoperative SVA and PJF, and their interactions were identified using a machine learning algorithm and game theory. Clinical benefits will depend on patient risk profiles.

4.
Nat Cancer ; 4(3): 419-435, 2023 03.
Article in English | MEDLINE | ID: mdl-36973439

ABSTRACT

Most tumor cells undergo apoptosis in circulation and at the metastatic organ sites due to host immune surveillance and a hostile microenvironment. It remains to be elucidated whether dying tumor cells have a direct effect on live tumor cells during the metastatic process and what the underlying mechanisms are. Here we report that apoptotic cancer cells enhance the metastatic outgrowth of surviving cells through Padi4-mediated nuclear expulsion. Tumor cell nuclear expulsion results in an extracellular DNA-protein complex that is enriched with receptor for advanced glycation endproducts (RAGE) ligands. The chromatin-bound RAGE ligand S100a4 activates RAGE receptors in neighboring surviving tumor cells, leading to Erk activation. In addition, we identified nuclear expulsion products in human patients with breast, bladder and lung cancer and a nuclear expulsion signature correlated with poor prognosis. Collectively, our study demonstrates how apoptotic cell death can enhance the metastatic outgrowth of neighboring live tumor cells.


Subject(s)
Lung Neoplasms , S100 Calcium-Binding Protein A4 , Humans , Apoptosis , Lung Neoplasms/metabolism , Receptor for Advanced Glycation End Products/metabolism , S100 Calcium-Binding Protein A4/genetics , S100 Calcium-Binding Protein A4/metabolism , Tumor Microenvironment
5.
Int J Mol Sci ; 23(16)2022 Aug 18.
Article in English | MEDLINE | ID: mdl-36012586

ABSTRACT

Triple-negative breast cancer (TNBC) is aggressive and has a poor overall survival due to a lack of therapeutic targets compared to other subtypes. Chemokine signature revealed that TNBC had low levels of CXCL14, an orphan homeostatic chemokine to regulate the immune network. Here, we investigated if CXCL14 plays a critical role in TNBC progression, focusing on survival rates, tumor growth and metastasis, and immune profiles in the tumor microenvironment. Analysis of human breast-cancer datasets showed that low CXCL14 expression levels were associated with poor survival rates in patients with breast cancer, particularly for TNBC subtypes. Overexpression of CXCL14 in TNBC 4T1 orthotopic mouse model significantly reduced tumor weights and inhibited lung metastasis. Furthermore, the CXCL14 overexpression altered immune profiles in the tumor microenvironment as follows: decreased F4/80+ macrophages and CD4+CD25+ Treg cells, and increased CD8+T cells in primary tumors; decreased Ly6C+ myeloid cells and CD4+CD25+ Treg cells and increased CD4+ and CD8+T cells in lung metastatic tumors. CXCL14-induced reduction of tumor growth and metastasis was diminished in T cell-deficient nude mice. Taken together, our data demonstrate that CXCL14 inhibits TNBC progression through altering immune profiles in the tumor microenvironment and it is mediated in a T cell-dependent manner. Thus, CXCL14 could be used as a biomarker for prognosis.


Subject(s)
CD8-Positive T-Lymphocytes , Chemokines, CXC , T-Lymphocytes, Regulatory , Triple Negative Breast Neoplasms , Animals , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Humans , Mice , Mice, Nude , T-Lymphocytes, Regulatory/immunology , Triple Negative Breast Neoplasms/pathology , Tumor Microenvironment
6.
Pharmacol Ther ; 237: 108253, 2022 09.
Article in English | MEDLINE | ID: mdl-35872332

ABSTRACT

Triple-negative breast cancer (TNBC) is an aggressive subtype characterized by extensive intra-tumoral heterogeneity, and frequently develops resistance to therapies. Tumor heterogeneity and lack of biomarkers are thought to be some of the most difficult challenges driving therapeutic resistance and relapse. This review will summarize current therapy for TNBC, studies in treatment resistance and relapse, including data from recent single cell sequencing. We will discuss changes in both the transcriptome and epigenome of TNBC, and we will review mechanisms regulating the immune microenvironment. Lastly, we will provide new perspective in patient stratification, and treatment options targeting transcriptome dysregulation and the immune microenvironment of TNBC patients.


Subject(s)
Triple Negative Breast Neoplasms , Biomarkers, Tumor , Humans , Neoplasm Recurrence, Local/genetics , Transcriptome , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/genetics , Tumor Microenvironment/genetics
7.
Mol Cancer Res ; 20(11): 1674-1685, 2022 11 03.
Article in English | MEDLINE | ID: mdl-35857355

ABSTRACT

The mechanisms of how cancer cells are selected and evolve to establish distant metastatic colonies remain unclear. Tumor heterogeneity and lack of biomarkers are some of the most difficult challenges in cancer biology and treatment. Here using mouse models for triple-negative breast cancer (TNBC) metastasis, we report heterogeneous expression of DNA methyltransferase 3B (DNMT3B) in both mouse and human primary tumors. High levels of DNMT3B were correlated with poor clinical outcomes in multiple human breast cancer datasets. Mechanistically, clonal cells with high DNMT3B (DNMT3BH) showed higher vimentin (VIM) expression and displayed enhanced epithelial-to-mesenchymal transition capacity. Deletion of VIM diminished the metastatic phenotype of DNMT3BH cells. Importantly, in preclinical mouse models in which the primary tumors were surgically removed, perioperative targeting of DNMT3B in combination with chemotherapy markedly suppressed tumor recurrence and metastasis. Our studies identify DNMT3B-mediated transcription regulation as an important mediator of tumor heterogeneity and show that DNMT3B is critical for tumor invasion and metastasis, reinforcing its potential as a target for treating metastatic disease. IMPLICATIONS: Our findings of transcriptome changes mediated by DNMT3B provide new mechanistic insight for intratumor heterogeneity and chemoresistance, and therapeutic targeting of DNMT3B in combination with chemotherapy offer additional treatment options for metastatic disease especially for patients with TNBC.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases , Triple Negative Breast Neoplasms , Animals , Humans , Mice , Cell Line, Tumor , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic , Neoplasm Recurrence, Local/genetics , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/metabolism , DNA Methyltransferase 3B
8.
Cancer Res ; 80(12): 2612-2627, 2020 06 15.
Article in English | MEDLINE | ID: mdl-32265226

ABSTRACT

Current cancer treatments are largely based on the genetic characterization of primary tumors and are ineffective for metastatic disease. Here we report that DNA methyltransferase 3B (DNMT3B) is induced at distant metastatic sites and mediates epigenetic reprogramming of metastatic tumor cells. Multiomics analysis and spontaneous metastatic mouse models revealed that DNMT3B alters multiple pathways including STAT3, NFκB, PI3K/Akt, ß-catenin, and Notch signaling, which are critical for cancer cell survival, apoptosis, proliferation, invasion, and colonization. PGE2 and IL6 were identified as critical inflammatory mediators in DNMT3B induction. DNMT3B expression levels positively correlated with human metastatic progression. Targeting IL6 or COX-2 reduced DNMT3B induction and improved chemo or PD1 therapy. We propose a novel mechanism linking the metastatic microenvironment with epigenetic alterations that occur at distant sites. These results caution against the "Achilles heel" in cancer therapies based on primary tumor characterization and suggests targeting DNMT3B induction as new option for treating metastatic disease. SIGNIFICANCE: These findings reveal that DNMT3B epigenetically regulates multiple pro-oncogenic signaling pathways via the inflammatory microenvironment at distant sites, cautioning the clinical approach basing current therapies on genetic characterization of primary tumors.


Subject(s)
Breast Neoplasms/pathology , DNA (Cytosine-5-)-Methyltransferases/metabolism , Dinoprostone/metabolism , Interleukin-6/metabolism , Lung Neoplasms/secondary , Animals , Antineoplastic Agents, Immunological/pharmacology , Antineoplastic Agents, Immunological/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast/pathology , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Cell Line, Tumor/transplantation , Cyclooxygenase 2/metabolism , Cyclooxygenase 2 Inhibitors/pharmacology , Cyclooxygenase 2 Inhibitors/therapeutic use , Datasets as Topic , Disease Models, Animal , Disease Progression , Epigenesis, Genetic/drug effects , Epigenesis, Genetic/immunology , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/immunology , Gene Knockdown Techniques , Humans , Interleukin-6/antagonists & inhibitors , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Mice , Programmed Cell Death 1 Receptor/immunology , Proof of Concept Study , Signal Transduction/drug effects , Signal Transduction/genetics , Signal Transduction/immunology , Tumor Microenvironment/drug effects , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology , DNA Methyltransferase 3B
9.
Nat Commun ; 9(1): 2611, 2018 07 04.
Article in English | MEDLINE | ID: mdl-29973593

ABSTRACT

Tumor-derived soluble factors promote the production of Gr-1+CD11b+ immature myeloid cells, and TGFß signaling is critical in their immune suppressive function. Here, we report that miR-130a and miR-145 directly target TGFß receptor II (TßRII) and are down-regulated in these myeloid cells, leading to increased TßRII. Ectopic expression of miR-130a and miR-145 in the myeloid cells decreased tumor metastasis. This is mediated through a downregulation of type 2 cytokines in myeloid cells and an increase in IFNγ-producing cytotoxic CD8 T lymphocytes. miR-130a- and miR-145-targeted molecular networks including TGFß and IGF1R pathways were correlated with higher tumor stages in cancer patients. Lastly, miR-130a and miR-145 mimics, as well as IGF1R inhibitor NT157 improved anti-tumor immunity and inhibited metastasis in preclinical mouse models. These results demonstrated that miR-130a and miR-145 can reprogram tumor-associated myeloid cells by altering the cytokine milieu and metastatic microenvironment, thus enhancing host antitumor immunity.


Subject(s)
Gene Expression Regulation, Neoplastic , Immunity, Innate/drug effects , Lung Neoplasms/genetics , Mammary Neoplasms, Experimental/genetics , MicroRNAs/genetics , Receptor, Transforming Growth Factor-beta Type II/genetics , Animals , Antineoplastic Agents/pharmacology , CD11b Antigen/genetics , CD11b Antigen/immunology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Cell Line, Tumor , Drug Evaluation, Preclinical , Female , Humans , Injections, Intravenous , Interferon-gamma/genetics , Interferon-gamma/immunology , Lung Neoplasms/immunology , Lung Neoplasms/secondary , Lung Neoplasms/therapy , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/pathology , Mammary Neoplasms, Experimental/therapy , Mice , Mice, Transgenic , MicroRNAs/immunology , Myeloid Cells/drug effects , Myeloid Cells/immunology , Myeloid Cells/pathology , Oligoribonucleotides/administration & dosage , Oligoribonucleotides/genetics , Oligoribonucleotides/metabolism , Pyrogallol/analogs & derivatives , Pyrogallol/pharmacology , Receptor, IGF Type 1/antagonists & inhibitors , Receptor, IGF Type 1/genetics , Receptor, IGF Type 1/immunology , Receptor, Transforming Growth Factor-beta Type II/immunology , Receptors, Chemokine/genetics , Receptors, Chemokine/immunology , Signal Transduction , Sulfonamides/pharmacology , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/immunology
10.
J Steroid Biochem Mol Biol ; 173: 69-74, 2017 10.
Article in English | MEDLINE | ID: mdl-27650654

ABSTRACT

Gemini analogs of calcitriol, characterized by the extension of the C21-methyl group of calcitriol with a second chain, act as agonists of the vitamin D receptor (VDR). This second side chain of gemini is accommodated in a new cavity inside the VDR created by the structural rearrangement of the protein core. The resulting conformational change preserves the active state of the receptor and bestows gemini compounds with biological activities that exceed those of calcitriol. Of particular interest are gemini's anti-cancer properties, and in this study we demonstrate anti-proliferative and tumor-reducing abilities of BXL0124 and BXL0097, differing only by the presence or absence, respectively, of the methylene group on the A ring. BXL0124 acts as a more potent VDR agonist than its 19-nor counterpart by activating VDR-mediated transcription at lower concentrations. In a similar manner, BXL0124 is more active than BXL0097 in growth inhibition of breast cancer cells and reduction of tumor volume. Structural comparisons of BXL0097 and BXL0124, as their VDR complexes, explain the elevated activity of the latter.


Subject(s)
Breast Neoplasms/drug therapy , Calcitriol/analogs & derivatives , Vitamins/chemistry , Vitamins/therapeutic use , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Calcitriol/chemistry , Calcitriol/pharmacology , Calcitriol/therapeutic use , Cell Proliferation/drug effects , Female , HEK293 Cells , Humans , MCF-7 Cells , Mice, SCID , Molecular Docking Simulation , Receptors, Calcitriol/metabolism , Transcriptional Activation/drug effects , Vitamins/pharmacology
11.
Cancer Prev Res (Phila) ; 8(9): 807-16, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26130252

ABSTRACT

This study evaluated the anticancer activity and mechanism of action of a γ-tocopherol-rich tocopherol mixture, γ-TmT, in two different animal models of estrogen-induced breast cancer. The chemopreventive effect of γ-TmT at early (6 weeks), intermediate (18 weeks), and late (31 weeks) stages of mammary tumorigenesis was determined using the August-Copenhagen Irish rat model. Female rats receiving 17ß-estradiol (E2) implants were administered with different doses (0%, 0.05%, 0.1%, 0.3%, and 0.5%) of γ-TmT diet. Treatment with 0.3% and 0.5% γ-TmT decreased tumor volume and multiplicity. At 31 weeks, serum concentrations of E2 were significantly decreased by γ-TmT. γ-TmT preferentially induced expression of the E2-metabolizing enzyme CYP1A1, over CYP1B1 in the rat mammary tissues. Nrf2-dependent antioxidant response was stimulated by γ-TmT, as evident from enhanced expression of its downstream targets, NQO1, GCLM, and HMOX1. Serum concentrations of the oxidative stress marker, 8-isoprostane, were also decreased in the γ-TmT-treated groups. Treatment with γ-TmT increased expression of PPARγ and its downstream genes, PTEN and p27, whereas the cell proliferation marker, PCNA, was significantly reduced in γ-TmT-treated mammary tumors. In an orthotopic model in which human MCF-7 breast cancer cells were injected into the mammary fat pad of immunodeficient mice, γ-TmT inhibited E2-dependent tumor growth at all the doses tested. In conclusion, γ-TmT reduced mammary tumor development, in part through decreased E2 availability and reduced oxidative stress in mammary tissues; γ-TmT could thus be an effective agent for the prevention and treatment of E2-induced breast cancer.


Subject(s)
Antioxidants/metabolism , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Estrogens/metabolism , PPAR gamma/metabolism , PTEN Phosphohydrolase/metabolism , Proliferating Cell Nuclear Antigen/metabolism , gamma-Tocopherol/therapeutic use , Animals , Cell Line, Tumor , Dinoprost/analogs & derivatives , Dinoprost/chemistry , Estradiol/administration & dosage , Female , Gene Expression Regulation, Neoplastic , Humans , Immunoenzyme Techniques , Immunohistochemistry , MCF-7 Cells , Mammary Glands, Animal/metabolism , Mammary Neoplasms, Animal/prevention & control , Mammary Neoplasms, Experimental/prevention & control , Mice , Mice, Nude , Neoplasm Transplantation , Oxidative Stress , RNA, Messenger/metabolism , Rats , Time Factors
12.
J Steroid Biochem Mol Biol ; 148: 148-55, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25445919

ABSTRACT

Breast cancer stem cells (BCSCs) are a subset of tumor cells that are believed to be the cells responsible for the establishment and maintenance of tumors. Moreover, BCSCs are suggested to be the main cause of progression to metastasis and recurrence of cancer because of their tumor-initiating abilities and resistance to conventional therapies. Ductal carcinoma in situ (DCIS) is an early precursor in breast carcinogenesis which progresses to invasive ductal carcinoma (IDC). We have previously reported that a vitamin D compound, BXL0124, inhibits the progression of DCIS to IDC. In the present study we sought to determine whether this effect was mediated through an influence on BCSCs. In MCF10DCIS cells treated with vitamin D compounds (1α25(OH)2D3 or BXL0124), the breast cancer stem cell-like population, identified by the CD44(+)/CD24(-/low) and CD49f(+)/CD24(-/low) subpopulations, was reduced. To determine the effects of vitamin D compounds on cancer stem cell activity, the MCF10DCIS mammosphere cell culture system, which enriches for mammary progenitor cells and putative BCSCs, was utilized. Untreated MCF10DCIS mammospheres showed a disorganized and irregular shape. When MCF10DCIS cells were treated with 1α25(OH)2D3 or BXL0124, the mammospheres that formed exhibited a more organized, symmetrical and circular shape, similar to the appearance of spheres formed by the non-malignant, normal mammary epithelial cell line, MCF10A. The mammosphere forming efficiency (MFE) was significantly decreased upon treatment with 1α25(OH)2D3 or BXL0124, indicating that these compounds have an inhibitory effect on mammosphere development. Treatment with 1α25(OH)2D3 or BXL0124 repressed markers associated with the stem cell-like phenotype, such as CD44, CD49f, c-Notch1, and pNFκB. Furthermore, 1α25(OH)2D3 and BXL0124 reduced the expression of pluripotency markers, OCT4 and KLF-4 in mammospheres. This study suggests that vitamin D compounds repress the breast cancer stem cell-like population, potentially contributing to their inhibition of breast cancer. This article is part of a Special Issue entitled '17th Vitamin D Workshop'.


Subject(s)
Biomarkers, Tumor/metabolism , Breast Neoplasms/drug therapy , Breast/drug effects , Neoplastic Stem Cells/drug effects , Vitamin D/pharmacology , Vitamins/pharmacology , Apoptosis/drug effects , Biomarkers, Tumor/genetics , Blotting, Western , Breast/metabolism , Breast/pathology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Female , Flow Cytometry , Humans , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Spheroids, Cellular/drug effects , Tumor Cells, Cultured
13.
J Steroid Biochem Mol Biol ; 148: 79-85, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25460302

ABSTRACT

Cancer stem cells (CSCs) are a small subset of cells that may be responsible for initiation, progression, and recurrence of tumors. Recent studies have demonstrated that CSCs are highly tumorigenic and resistant to conventional chemotherapies, making them a promising target for the development of preventive/therapeutic agents. A single or combination of various markers, such as CD44, EpCAM, CD49f, CD133, CXCR4, ALDH-1, and CD24, were utilized to isolate CSCs from various types of human cancers. Notch, Hedgehog, Wnt, and TGF-ß signalingregulate self-renewal and differentiation of normal stem cells andare aberrantly activated in CSCs. In addition, many studies have demonstrated that these stem cell-associated signaling pathways are required for the maintenance of CSCs in different malignancies, including breast, colorectal, prostate, and pancreatic cancers. Accumulating evidence has shown inhibitory effects of vitamin D and its analogs on the cancer stem cell signaling pathways, suggesting vitamin D as a potential preventive/therapeutic agent against CSCs. In this review, we summarize recent findings about the roles of Notch, Hedgehog, Wnt, and TGF-ß signaling in CSCs as well as the effects of vitamin D on these stem cell signaling pathways. This article is part of a Special Issue entitled '17th Vitamin D Workshop'.


Subject(s)
Neoplasms/drug therapy , Neoplasms/pathology , Neoplastic Stem Cells/drug effects , Vitamin D/pharmacology , Vitamins/pharmacology , Animals , Humans , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology
14.
J Steroid Biochem Mol Biol ; 148: 111-21, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25541438

ABSTRACT

Tumor-initiating cells (also known as cancer stem cells) are the subpopulation of cells shown to be responsible for tumor initiation, maintenance and recurrence. In breast cancer, CD44(+)/CD24(-/low) cells were identified as tumor-initiating cells. We previously reported that a Gemini vitamin D analog, 1,25-dihydroxy-20R-21(3-hydroxy-3-deuteromethyl-4,4,4-trideuterobutyl)-23-yne-26,27-hexafluoro-cholecalciferol (BXL0124), reduced CD44(+)/CD24(-/low) cells in MCF10DCIS basal-like breast cancer cells. Since Notch has been identified as one of the key signaling pathways involved in breast cancer stem cells, the effect of BXL0124 on the Notch signaling pathway was investigated in breast cancer. The CD44(+)/CD24(-/low) subpopulation of MCF10DCIS cells showed elevated Notch1 signaling and increased cell proliferation compared to the CD44(+)/CD24(high) subpopulation. Treatment with the Gemini vitamin D analog BXL0124 decreased the level of activated Notch1 receptor. In addition, mRNA and protein levels of the Notch ligands, Jagged-1, Jagged-2 and DLL1, were significantly reduced by treatment with BXL0124, which was followed by repression of c-Myc, a key downstream target of Notch signaling. Interestingly, HES1, a known downstream target of Notch signaling, was rapidly induced by treatment with BXL0124. The inhibitory effect of BXL0124 on Notch signaling was reversed by knockdown of HES1. Overexpression of HES1 inhibited Notch1 signaling and reduced the CD44(+)/CD24(-/low) subpopulation, confirming a role of HES1 in Notch1 signaling. In conclusion, the Gemini vitamin D analog, BXL0124, represses the tumor-initiating subpopulation by HES1-mediated inhibition of Notch1 signaling. The present study demonstrates BXL0124 as a potent inhibitor of Notch signaling to target tumor-initiating cells in basal-like breast cancer. This article is part of a Special Issue entitled "17th Vitamin D Workshop".


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Breast Neoplasms/drug therapy , CD24 Antigen/metabolism , Calcitriol/analogs & derivatives , Carcinoma, Basal Cell/drug therapy , Homeodomain Proteins/metabolism , Hyaluronan Receptors/metabolism , Neoplastic Stem Cells/drug effects , Receptor, Notch1/metabolism , Apoptosis/drug effects , Basic Helix-Loop-Helix Transcription Factors/antagonists & inhibitors , Basic Helix-Loop-Helix Transcription Factors/genetics , Blotting, Western , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Calcitriol/pharmacology , Carcinoma, Basal Cell/metabolism , Carcinoma, Basal Cell/pathology , Cell Proliferation/drug effects , Female , Flow Cytometry , Homeodomain Proteins/antagonists & inhibitors , Homeodomain Proteins/genetics , Humans , Microscopy, Fluorescence , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Receptor, Notch1/antagonists & inhibitors , Receptor, Notch1/genetics , Receptors, Calcitriol/antagonists & inhibitors , Receptors, Calcitriol/genetics , Receptors, Calcitriol/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factor HES-1 , Tumor Cells, Cultured
15.
Mol Carcinog ; 54(9): 916-25, 2015 Sep.
Article in English | MEDLINE | ID: mdl-24782330

ABSTRACT

Oxidative stress is known to play a key role in estrogen-induced breast cancer. This study assessed the chemopreventive activity of the naturally occurring γ-tocopherol-rich mixture of tocopherols (γ-TmT) in early stages of estrogen-induced mammary hyperplasia in ACI rats. ACI rats provide an established model of rodent mammary carcinogenesis due to their high sensitivity to estrogen. Female rats were implanted with 9 mg of 17ß-estradiol (E2) in silastic tubings and fed with control or 0.3% γ-TmT diet for 1, 3, 7, and 14 d. γ-TmT increased the levels of tocopherols and their metabolites in the serum and mammary glands of the rats. Histological analysis revealed mammary hyperplasia in the E2 treated rats fed with control or γ-TmT diet. γ-TmT decreased the levels of E2-induced nitrosative and oxidative stress markers, nitrotyrosine, and 8-oxo-dG, respectively, in the hyperplastic mammary tissues. 8-Isoprostane, a marker of oxidative stress in the serum, was also reduced by γ-TmT. Noticeably, γ-TmT stimulated Nrf2-dependent antioxidant response in the mammary glands of E2 treated rats, evident from the induced mRNA levels of Nrf2 and its downstream antioxidant enzymes, superoxide dismutase, catalase, and glutathione peroxidase. Therefore, inhibition of nitrosative/oxidative stress through induction of antioxidant response is the primary effect of γ-TmT in early stages of E2-induced mammary hyperplasia. Due to its cytoprotective activity, γ-TmT could be a potential natural agent for the chemoprevention of estrogen-induced breast cancer.


Subject(s)
Antioxidants/therapeutic use , Breast Diseases/diet therapy , Dietary Supplements , Mammary Glands, Animal/pathology , Oxidative Stress/drug effects , Tocopherols/therapeutic use , Animals , Breast Diseases/chemically induced , Breast Diseases/metabolism , Breast Diseases/pathology , Dietary Supplements/analysis , Estrogens , Female , Humans , Hyperplasia/chemically induced , Hyperplasia/diet therapy , Hyperplasia/metabolism , Hyperplasia/pathology , Mammary Glands, Animal/metabolism , NF-E2-Related Factor 2/genetics , RNA, Messenger/genetics , Rats , Rats, Inbred ACI , Tyrosine/analogs & derivatives , Tyrosine/analysis , Up-Regulation
16.
PLoS One ; 9(9): e107616, 2014.
Article in English | MEDLINE | ID: mdl-25229616

ABSTRACT

Triple-negative breast cancer is associated with poor prognosis because of a high rate of tumor recurrence and metastasis. Previous studies demonstrated that the synthetic triterpenoid, CDDO-Imidazolide (CDDO-Im) induced cell cycle arrest and apoptosis in triple-negative breast cancer. Since a small subpopulation of cancer stem cells has been suggested to be responsible for drug resistance and metastasis of tumors, our present study determined whether the effects of CDDO-Im in triple-negative breast cancer are due to the inhibition of a cancer stem cell subpopulation. CDDO-Im treatment markedly induced cell cycle arrest at G2/M-phase and apoptosis in the triple-negative breast cancer cell lines, SUM159 and MDA-MB-231. Because SUM159 cells were more sensitive to CDDO-Im than MDA-MB-231 cells, the effects of CDDO-Im on the cancer stem cell subpopulation were further investigated in SUM159 cells. SUM159 cells formed tumorspheres in culture, and the cancer stem cell subpopulation, CD24-/EpCAM+ cells, was markedly enriched in SUM159 tumorspheres. The CD24-/EpCAM+ cells in SUM159 tumorspheres were significantly inhibited by CDDO-Im treatment. CDDO-Im also significantly decreased sphere forming efficiency and tumorsphere size in both primary and secondary sphere cultures. PCR array of stem cell signaling genes showed that expression levels of many key molecules in the stem cell signaling pathways, such as Notch, TGF-ß/Smad, Hedgehog and Wnt, were significantly down-regulated by CDDO-Im in SUM159 tumorspheres. Protein levels of Notch receptors (c-Notch1, Notch1 and Notch3), TGF-ß/Smad (pSmad2/3) and Hedgehog downstream effectors (GLI1) also were markedly reduced by CDDO-Im. In conclusion, the present study demonstrates that the synthetic triterpenoid, CDDO-Im, is a potent anti-cancer agent against triple-negative breast cancer cells by targeting the cancer stem cell subpopulation.


Subject(s)
Antineoplastic Agents/pharmacology , Imidazoles/pharmacology , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Oleanolic Acid/analogs & derivatives , Signal Transduction/drug effects , Triple Negative Breast Neoplasms/pathology , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Dose-Response Relationship, Drug , G2 Phase Cell Cycle Checkpoints/drug effects , Hedgehog Proteins/metabolism , Humans , M Phase Cell Cycle Checkpoints/drug effects , Oleanolic Acid/pharmacology , Receptors, Notch/metabolism , Transforming Growth Factor beta/metabolism
17.
Cancer Prev Res (Phila) ; 7(6): 617-26, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24691501

ABSTRACT

Ductal carcinoma in situ (DCIS) is a nonmalignant lesion of the breast with the potential to progress to invasive ductal carcinoma (IDC). The disappearance and breakdown of the myoepithelial cell layer and basement membrane in DCIS have been identified as major events in the development of breast cancer. The MCF10DCIS.com cell line is a well-established model, which recapitulates the progression of breast cancer from DCIS to IDC. We have previously reported that a novel Gemini vitamin D analog, 1α,25-dihydroxy-20R-21(3-hydroxy-3-deuteromethyl-4,4,4-trideuterobutyl)-23-yne-26,27-hexafluoro-cholecalciferol (BXL0124) is a potent inhibitor of the growth of MCF10DCIS.com xenografted tumors without hypercalcemic toxicity. In this study, we utilized the MCF10DCIS.com in vivo model to assess the effects of BXL0124 on breast cancer progression from weeks 1 to 4. Upon DCIS progression to IDC from weeks 3 to 4, tumors lost the myoepithelial cell layer and basement membrane as shown by immunofluorescence staining with smooth muscle actin and laminin 5, respectively. Administration of BXL0124 maintained the critical myoepithelial cell layer as well as basement membrane, and animals treated with BXL0124 showed a 43% reduction in tumor volume by week 4. BXL0124 treatment decreased cell proliferation and maintained vitamin D receptor levels in tumors. In addition, the BXL0124 treatment reduced the mRNA levels of matrix metalloproteinases starting at week 3, contributing to the inhibition of invasive transition. Our results suggest that the maintenance of DCIS plays a significant role in the cancer preventive action of the Gemini vitamin D BXL0124 during the progression of breast lesions.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Calcitriol/analogs & derivatives , Carcinoma, Ductal, Breast/prevention & control , Carcinoma, Intraductal, Noninfiltrating/drug therapy , Carcinoma, Intraductal, Noninfiltrating/pathology , Epithelial-Mesenchymal Transition/drug effects , Animals , Calcitriol/chemistry , Calcitriol/therapeutic use , Carcinoma, Ductal, Breast/pathology , Cell Proliferation/drug effects , Down-Regulation/drug effects , Female , Humans , Mice , Mice, Nude , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
18.
Cancer Prev Res (Phila) ; 6(9): 959-70, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23856074

ABSTRACT

HER2 (or ErbB2), a member of ErbB receptor tyrosine kinases, is overexpressed in approximately 20% of human breast cancer, and the ErbB2 signaling pathway is a critical therapeutic target for ErbB2-overexpressing breast cancer. We investigated the inhibitory effects of the Gemini vitamin D analog BXL0124, the synthetic triterpenoid CDDO-Im and the combination on the tumorigenesis of ErbB2-overexpressing breast cancer. MMTV-ErbB2/neu transgenic mice were treated with BXL0124, CDDO-Im, or the combination from three months of age until the end of the experiment. Formation and growth of MMTV-ErbB2/neu mammary tumors were monitored every week, and all three treatments delayed the development of mammary tumors without significant toxicity. Decreased activation of ErbB2 as well as other ErbB receptors, ErbB1 and ErbB3, in MMTV-ErbB2/neu mammary tumors was shown by all treatments. Protein levels of downstream targets of the ErbB2 signaling pathway, including activated-Erk1/2, activated-Akt, c-Myc, CycD1, and Bcl2, were repressed by all three treatments, with the combination treatment exhibiting the strongest effects. To investigate therapeutic efficacy, the combination of BXL0124 and CDDO-Im was given to MMTV-ErbB2/neu mice after mammary tumors were established between 23 and 30 weeks of age. Short-term treatment with the combination did not show effects on tumor growth nor the ErbB2 signaling pathway. The present study shows BXL0124, CDDO-Im, and the combination as potential agents for prevention, but not treatment, against the tumorigenesis of ErbB2-overexpressing breast cancer.


Subject(s)
Calcitriol/analogs & derivatives , Cell Transformation, Neoplastic/drug effects , Imidazoles/administration & dosage , Mammary Neoplasms, Animal/prevention & control , Oleanolic Acid/analogs & derivatives , Receptor, ErbB-2/physiology , Signal Transduction/drug effects , Administration, Oral , Animals , Antineoplastic Combined Chemotherapy Protocols , Blotting, Western , Calcitriol/administration & dosage , Calcitriol/pharmacology , Cell Transformation, Neoplastic/pathology , Female , Humans , Imidazoles/pharmacology , Mammary Neoplasms, Animal/metabolism , Mammary Neoplasms, Animal/pathology , Mammary Tumor Virus, Mouse/genetics , Mice , Mice, Transgenic , Microscopy, Fluorescence , Oleanolic Acid/administration & dosage , Oleanolic Acid/pharmacology , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
19.
PLoS One ; 8(1): e54020, 2013.
Article in English | MEDLINE | ID: mdl-23326564

ABSTRACT

BACKGROUND: CD44, a transmembrane glycoprotein, is a major receptor for extracellular proteins involved in invasion and metastasis of human cancers. We have previously demonstrated that the novel Gemini vitamin D analog BXL0124 [1α,25-dihydroxy-20R-21(3-hydroxy-3-deuteromethyl-4,4,4-trideuterobutyl)-23-yne-26,27-hexafluro-cholecalciferol] repressed CD44 expression in MCF10DCIS.com basal-like human breast cancer cells and inhibited MCF10DCIS xenograft tumor growth. In the present study, we investigated potential factors downstream of CD44 and the biological role of CD44 repression by BXL0124 in MCF10DCIS cells. METHODS AND FINDINGS: The treatment with Gemini vitamin D BXL0124 decreased CD44 protein level, suppressed STAT3 signaling, and inhibited invasion and proliferation of MCF10DCIS cells. The interaction between CD44 and STAT3 was determined by co-immunoprecipitation. CD44 forms a complex with STAT3 and Janus kinase 2 (JAK2) to activate STAT3 signaling, which was inhibited by BXL0124 in MCF10DCIS cells. The role of CD44 in STAT3 signaling and invasion of MCF10DCIS cells was further determined by the knockdown of CD44 using small hairpin RNA in vitro and in vivo. MCF10DCIS cell invasion was markedly decreased by the knockdown of CD44 in vitro. The knockdown of CD44 also significantly decreased mRNA expression levels of invasion markers, matrix metalloproteinases (MMPs) and urokinase plasminogen activator (uPA), in MCF10DCIS cells. In MCF10DCIS xenograft tumors, CD44 knockdown decreased tumor size and weight as well as invasion markers. CONCLUSIONS: The present study identifies STAT3 as an important signaling molecule interacting with CD44 and demonstrates the essential role of CD44-STAT3 signaling in breast cancer invasion. It also suggests that repression of CD44-STAT3 signaling is a key molecular mechanism in the inhibition of breast cancer invasion by the Gemini vitamin D analog BXL0124.


Subject(s)
Breast Neoplasms , Gene Expression Regulation, Neoplastic/drug effects , Hyaluronan Receptors/metabolism , STAT3 Transcription Factor/metabolism , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Calcitriol/analogs & derivatives , Calcitriol/pharmacology , Cell Line, Tumor , Female , Humans , Hyaluronan Receptors/genetics , Mice , Neoplasm Invasiveness , Neoplasm Metastasis/drug therapy , STAT3 Transcription Factor/genetics , Signal Transduction , Transplantation, Heterologous , Vitamin D/analogs & derivatives , Vitamin D/pharmacology
20.
Mol Carcinog ; 52(7): 514-25, 2013 Jul.
Article in English | MEDLINE | ID: mdl-22389237

ABSTRACT

Previous clinical and epidemiological studies of vitamin E have used primarily α-tocopherol for the prevention of cancer. However, γ-tocopherol has demonstrated greater anti-inflammatory and anti-tumor activity than α-tocopherol in several animal models of cancer. This study assessed the potential chemopreventive activities of a tocopherol mixture containing 58% γ-tocopherol (γ-TmT) in an established rodent model of mammary carcinogenesis. Female ACI rats were utilized due to their sensitivity to 17ß-estradiol (E2 ) to induce mammary hyperplasia and neoplasia. The rats were implanted subcutaneously with sustained release E2 pellets and given dietary 0.3% or 0.5% γ-TmT for 2 or 10 wk. Serum E2 levels were significantly reduced by the treatment with 0.5% γ-TmT. Serum levels of inflammatory markers, prostaglandin E2 and 8-isoprostane, were suppressed by γ-TmT treatment. Histology of mammary glands showed evidence of epithelial hyperplasia in E2 -treated rats. Immunohistochemical analysis of the mammary glands revealed a decrease in proliferating cell nuclear antigen (PCNA), cyclooxygenase-2 (COX-2), and estrogen receptor α (ERα), while there was an increase in cleaved-caspase 3, peroxisome proliferator-activated receptor γ (PPARγ), and nuclear factor (erythroid-derived 2)-like 2 (Nrf2) in γ-TmT-treated rats. In addition, treatment with γ-TmT resulted in a decrease in the expression of ERα mRNA, whereas mRNA levels of ERß and PPARγ were increased. In conclusion, γ-TmT was shown to suppress inflammatory markers, inhibit E2 -induced cell proliferation, and upregulate PPARγ and Nrf2 expression in mammary hyperplasia, suggesting that γ-TmT may be a promising agent for human breast cancer prevention.


Subject(s)
Cell Proliferation , Diet , Estrogen Receptor alpha/metabolism , Mammary Neoplasms, Experimental/prevention & control , NF-E2-Related Factor 2/metabolism , PPAR gamma/metabolism , Tocopherols/administration & dosage , Animals , Antioxidants/administration & dosage , Antioxidants/metabolism , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Blotting, Western , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Estradiol/blood , Estrogen Receptor alpha/genetics , Female , Gene Expression Regulation, Neoplastic , Hyperplasia/metabolism , Hyperplasia/pathology , Hyperplasia/prevention & control , Immunoenzyme Techniques , Inflammation Mediators/metabolism , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Microsomes, Liver/metabolism , NF-E2-Related Factor 2/genetics , PPAR gamma/genetics , RNA, Messenger/genetics , Rats , Rats, Inbred ACI , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Tocopherols/blood
SELECTION OF CITATIONS
SEARCH DETAIL
...