Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Sci Rep ; 13(1): 17597, 2023 10 16.
Article in English | MEDLINE | ID: mdl-37845346

ABSTRACT

The intestinal epithelium is highly regenerative. Rapidly proliferating LGR5+ crypt base columnar (CBC) cells are responsible for epithelial turnover needed to maintain intestinal homeostasis. Upon tissue damage, loss of LGR5+ CBCs can be compensated by activation of quiescent +4 intestinal stem cells (ISCs) or early progenitor cells to restore intestinal regeneration. LGR5+ CBC self-renewal and ISC conversion to LGR5+ cells are regulated by external signals originating from the ISC niche. In contrast, little is known about intrinsic regulatory mechanisms critical for maintenance of LGR5+ CBC homeostasis. We found that LGR5 expression in intestinal crypt cells is controlled by the circadian core clock gene BMAL1 and the BMAL1-regulated RNA-binding protein MEX3A. BMAL1 directly activated transcription of Mex3a. MEX3A in turn bound to and stabilized Lgr5 mRNA. Bmal1 depletion reduced Mex3a and Lgr5 expression and led to increased ferroptosis, which consequently decreased LGR5+ CBC numbers and increased the number of crypt cells expressing +4 ISC marker BMI1. Together, these findings reveal a BMAL1-centered intrinsic regulatory pathway that maintains LGR5 expression in the crypt cells and suggest a potential mechanism contributing to ISC homeostasis.


Subject(s)
ARNTL Transcription Factors , Intestines , ARNTL Transcription Factors/genetics , ARNTL Transcription Factors/metabolism , Intestinal Mucosa/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism
2.
Cancer Res ; 83(2): 251-263, 2023 01 18.
Article in English | MEDLINE | ID: mdl-36354374

ABSTRACT

Epithelial ovarian cancer is a highly heterogeneous and malignant female cancer with an overall low survival rate. Mutations in p53 are prevalent in the major ovarian cancer histotype, high-grade serous ovarian carcinoma (HGSOC), while p53 mutations are much less frequent in other ovarian cancer subtypes, particularly in ovarian clear cell carcinoma (OCCC). Advanced stage OCCC with wild-type (WT) p53 has a worse prognosis and increased drug resistance, metastasis, and recurrence than HGSOC. The mechanisms responsible for driving the aggressiveness of WT p53-expressing ovarian cancer remain poorly understood. Here, we found that upregulation of MEX3A, a dual-function protein containing a RING finger domain and an RNA-binding domain, was critical for tumorigenesis in WT p53-expressing ovarian cancer. MEX3A overexpression enhanced the growth and clonogenicity of OCCC cell lines. In contrast, depletion of MEX3A in OCCC cells, as well as ovarian teratocarcinoma cells, reduced cell survival and proliferative ability. MEX3A depletion also inhibited tumor growth and prolonged survival in orthotopic xenograft models. MEX3A depletion did not alter p53 mRNA level but did increase p53 protein stability. MEX3A-mediated p53 protein degradation was crucial to suppress ferroptosis and enhance tumorigenesis. Consistently, p53 knockdown reversed the effects of MEX3A depletion. Together, our observations identified MEX3A as an important oncogenic factor promoting tumorigenesis in ovarian cancer cells expressing WT p53. SIGNIFICANCE: Degradation of p53 mediated by MEX3A drives ovarian cancer growth by circumventing p53 tumor suppressive functions, suggesting targeting MEX3A as a potential strategy for treating of ovarian cancer expressing WT p53.


Subject(s)
Adenocarcinoma, Clear Cell , Ferroptosis , Ovarian Neoplasms , RNA-Binding Proteins , Tumor Suppressor Protein p53 , Female , Humans , Adenocarcinoma, Clear Cell/drug therapy , Carcinogenesis/genetics , Carcinoma, Ovarian Epithelial , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Ferroptosis/genetics , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Phosphoproteins/genetics , Phosphoproteins/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
3.
Sci Rep ; 11(1): 14431, 2021 07 13.
Article in English | MEDLINE | ID: mdl-34257372

ABSTRACT

Posttranslational modification (PTM) of core circadian clock proteins, including Period2 (PER2), is required for proper circadian regulation. PER2 function is regulated by casein kinase 1 (CK1)-mediated phosphorylation and ubiquitination but little is known about other PER2 PTMs or their interaction with PER2 phosphorylation. We found that PER2 can be SUMOylated by both SUMO1 and SUMO2; however, SUMO1 versus SUMO2 conjugation had different effects on PER2 turnover and transcriptional suppressor function. SUMO2 conjugation facilitated PER2 interaction with ß-TrCP leading to PER2 proteasomal degradation. In contrast, SUMO1 conjugation, mediated by E3 SUMO-protein ligase RanBP2, enhanced CK1-mediated PER2S662 phosphorylation, inhibited PER2 degradation and increased PER2 transcriptional suppressor function. PER2 K736 was critical for both SUMO1- and SUMO2-conjugation. A PER2K736R mutation was sufficient to alter PER2 protein oscillation and reduce PER2-mediated transcriptional suppression. Together, our data revealed that SUMO1 versus SUMO2 conjugation acts as a determinant of PER2 stability and function and thereby affects the circadian regulatory system and the expression of clock-controlled genes.


Subject(s)
SUMO-1 Protein , Animals , Circadian Rhythm , Genes, Regulator , Humans , Phosphorylation , Sumoylation , Ubiquitination
4.
Proc Natl Acad Sci U S A ; 118(3)2021 01 19.
Article in English | MEDLINE | ID: mdl-33431674

ABSTRACT

Metastasis is the major cause of cancer death. An increased level of circulating tumor cells (CTCs), metastatic cancer cells that have intravasated into the circulatory system, is particularly associated with colonization of distant organs and poor prognosis. However, the key factors required for tumor cell dissemination and colonization remain elusive. We found that high expression of desmoglein2 (DSG2), a component of desmosome-mediated intercellular adhesion complexes, promoted tumor growth, increased the prevalence of CTC clusters, and facilitated distant organ colonization. The dynamic regulation of DSG2 by hypoxia was key to this process, as down-regulation of DSG2 in hypoxic regions of primary tumors led to elevated epithelial-mesenchymal transition (EMT) gene expression, allowing cells to detach from the primary tumor and undergo intravasation. Subsequent derepression of DSG2 after intravasation and release of hypoxic stress was associated with an increased ability to colonize distant organs. This dynamic regulation of DSG2 was mediated by Hypoxia-Induced Factor1α (HIF1α). In contrast to its more widely observed function to promote expression of hypoxia-inducible genes, HIF1α repressed DSG2 by recruitment of the polycomb repressive complex 2 components, EZH2 and SUZ12, to the DSG2 promoter in hypoxic cells. Consistent with our experimental data, DSG2 expression level correlated with poor prognosis and recurrence risk in breast cancer patients. Together, these results demonstrated the importance of DSG2 expression in metastasis and revealed a mechanism by which hypoxia drives metastasis.


Subject(s)
Breast Neoplasms/genetics , Desmoglein 2/genetics , Epithelial-Mesenchymal Transition/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia/genetics , Neoplasm Recurrence, Local/genetics , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Cell Line, Tumor , Desmoglein 2/antagonists & inhibitors , Desmoglein 2/metabolism , Enhancer of Zeste Homolog 2 Protein/genetics , Enhancer of Zeste Homolog 2 Protein/metabolism , Female , Gene Expression Regulation, Neoplastic , Humans , Hypoxia/metabolism , Hypoxia/mortality , Hypoxia/pathology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Lymphatic Metastasis , Mice , Mice, SCID , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neoplasm Recurrence, Local/metabolism , Neoplasm Recurrence, Local/mortality , Neoplasm Recurrence, Local/pathology , Neoplastic Cells, Circulating/metabolism , Neoplastic Cells, Circulating/pathology , Polycomb Repressive Complex 2/genetics , Polycomb Repressive Complex 2/metabolism , Promoter Regions, Genetic , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Survival Analysis , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Burden , Xenograft Model Antitumor Assays
5.
Environ Toxicol ; 34(1): 73-82, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30259641

ABSTRACT

Breast cancer (BC) is the most common cancer affecting women worldwide and has been associated with active tobacco smoking. Low levels of nicotine (Nic) and 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK), have been detected in cases of second-hand smoke (SHS). However, the correlation between SHS and BC risk remains controversial. In this study, we investigated whether the physiological SHS achievable dose of Nic and tobacco specific nitrosamine, NNK act together to induce breast carcinogenesis using an in vitro breast cell carcinogenesis model. Immortalized non-tumorigenic breast epithelial cell line, HBL-100 used for a time-course assay, was exposed to very low levels of either Nic or NNK, or both. The time-course assay consisted of 23 cycles of nitrosamines treatment. In each cycle, HBL-100 cells were exposed to 1pM of Nic and/or 100 femtM of NNK for 48 hours. Cells were passaged every 3 days and harvested after 10, 15, and 23 cycles. Our results demonstrated that the tumorigenicity of HBL-100, defined by soft agar colony forming, proliferation, migration and invasion abilities, was enhanced by co-exposure to physiologically SHS achievable doses of Nic and NNK. In addition, α9-nAChR signaling activation, which plays an important role in cellular proliferation and cell survival, was also observed. Importantly, an increase in stemness properties including the prevalence of CD44+/CD24- cells, increase Nanog expression and mammosphere-forming ability were also observed. Our results indicate that chronic and long term exposure to environmental tobacco smoke, may induce breast cell carcinogenesis even at extremely low doses.


Subject(s)
Breast Neoplasms/chemically induced , Cell Transformation, Neoplastic/drug effects , Mammary Glands, Human/drug effects , Nicotine/toxicity , Nitrosamines/toxicity , Acetylcholine/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Carcinogens/toxicity , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cells, Cultured , Dose-Response Relationship, Drug , Epithelial Cells/drug effects , Epithelial Cells/physiology , Female , Humans , Mammary Glands, Human/pathology , Mammary Glands, Human/physiology , Receptors, Nicotinic/genetics , Receptors, Nicotinic/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Time Factors , Toxicity Tests, Chronic
6.
Biochim Biophys Acta Gene Regul Mech ; 1861(11): 1007-1017, 2018 11.
Article in English | MEDLINE | ID: mdl-30343691

ABSTRACT

Chronic inflammation is known to contribute to tumor initiation and cancer progression. In breast tissue, the core circadian gene Period (PER)2 plays a critical role in mammary gland development and possesses tumor suppressor function. Interleukin (IL)-6 and C-C motif chemokine ligand (CCL) 2 are among the most abundant cytokines in the inflammatory microenvironment. We found that acute stimulation by IL-6/CCL2 reduced PER2 expression in non-tumorigenic breast epithelial cells. Longer term exposure to IL-6/CCL2 suppressed PER2 to an even lower level. IL-6 activated STAT3/NFκB p50 signaling to recruit HDAC1 to the PER2 promoter. CCL2 activated the PI3K/AKT pathway to promote ELK-1 cytoplasm-to-nucleus translocation, recruit HDAC1 to the proximal PER2 promoter and facilitate DNMT3-EZH2-PER2 promoter association. Ectopic expression of PER2 inhibited IL-6 or CCL2 induced mammosphere forming ability and reduced sphere size indicating that PER2 repression in breast epithelial cells can be crucial to activate tumorigenesis in an inflammatory microenvironment. The diminished expression of PER2 can be observed over a time scale of hours to weeks following IL-6/CCL2 stimulation suggesting that PER2 suppression occurs in the early stage of the interaction between an inflammatory microenvironment and normal breast epithelial cells. These data show new mechanisms by which mammary cells interact with a cancerous microenvironment and provide additional evidence that PER2 expression contributes to breast tumorigenesis.


Subject(s)
Breast/cytology , Chemokine CCL2/physiology , Epithelial Cells/metabolism , Interleukin-6/physiology , Period Circadian Proteins/genetics , Breast Neoplasms/metabolism , Cells, Cultured , Down-Regulation , Female , Fibroblasts/metabolism , Gene Expression Regulation , Humans , NF-kappa B p50 Subunit/metabolism , Oncogene Protein v-akt/metabolism , Phosphatidylinositol 3-Kinases/metabolism , STAT3 Transcription Factor/metabolism
7.
Hum Pathol ; 80: 219-230, 2018 10.
Article in English | MEDLINE | ID: mdl-29902578

ABSTRACT

The transcription factor GATA3 plays a significant role in mammary gland development and differentiation. We analyzed expression of GATA3 in breast cancer (BC) cell lines and clinical specimens from BC patients in Taiwan. Semiquantitative reverse-transcription polymerase chain reaction (RT-PCR), quantitative real-time PCR were carried out to determine the mRNA level of GATA3 from 241 pairs of matched tumor and adjacent normal tissues from anonymous female donors. GATA3 immunohistochemistry (IHC) staining and H-score were performed (n = 25). Inducing and silencing of GATA3 were done by exposure MCF-7 cell line to nicotine or curcumin, respectively. GATA3 expression was detected in most of the estrogen receptor-positive (ER+) tumor specimens (176/241, 73%) compared with paired normal tissues (65/241, 27%) (P < .001). The GATA3 level was highest in Luminal A, and independent t-tests revealed higher GATA3 was associated with ER+ (P = .018) and BC stages (stage II, and stage IV). Nuclear protein expression of GATA3 was detected in tumor tissues (P < .001) with higher H-score in Luminal A patients (P = .012). Kaplan-Meier survival analyses showed that ER+/progesterone receptor (PgR)+ and lower grade BC patients with relatively high GATA3 had better clinical overall survival (OS). GATA3 regulates ERα and BCL-2 as BC luminal subtype markers. Cox univariate and multivariate analyses demonstrated that the expression of GATA3 was an effective predictor of the risk of death. We demonstrated a correlation between GATA3 expression and only ER+ and suggest that a higher GATA3 expression is a good prognostic factor for OS for ER+ BC patients.


Subject(s)
Biomarkers, Tumor/metabolism , Breast Neoplasms/pathology , GATA3 Transcription Factor/metabolism , Receptors, Progesterone/metabolism , Adult , Aged , Breast Neoplasms/diagnosis , Breast Neoplasms/mortality , Disease-Free Survival , Female , Humans , Kaplan-Meier Estimate , Middle Aged , Prognosis , Receptors, Estrogen/metabolism
8.
Front Pharmacol ; 9: 205, 2018.
Article in English | MEDLINE | ID: mdl-29593536

ABSTRACT

The combination cancer therapy is a new strategy to circumvent drug resistance for the treatment of high metastasis and advanced malignancies. Herein, we developed a synthesized compound MPT0B451 that display inhibitory effect against histone deacetylase (HDAC) 6 and tubulin assembly. Our data demonstrated that MPT0B451 significantly inhibited cancer cell growths in HL-60 and PC-3 cells due to inhibition of HDAC activity. MPT0B451 also markedly increased caspase-mediated apoptosis in these cells. The cell cycle analysis showed mitotic arrest induced by MPT0B451 with enhanced expression of G2/M transition proteins. Moreover, molecular docking analysis supported MPT0B451 as a dual HDAC6 and tubulin inhibitor. Finally, MPT0B451 led to tumor growth inhibition (TGI) in HL-60 and PC-3 xenograft models. These findings indicated that MPT0B451 has dual inhibition effects for HDAC6 and tubulin, and also contributed to G2/M arrest followed by apoptotic induction. Together, our results suggested that MPT0B451 may serve as a potent anti-cancer treatment regimen in human prostate cancer and acute myeloid leukemia.

9.
EMBO Mol Med ; 9(12): 1660-1680, 2017 12.
Article in English | MEDLINE | ID: mdl-28993429

ABSTRACT

Lymph node (LN) metastasis is commonly associated with systemic distant organ metastasis in human breast cancer and is an important prognostic predictor for survival of breast cancer patients. However, whether tumor-draining LNs (TDLNs) play a significant role in modulating the malignancy of cancer cells for distant metastasis remains controversial. Using a syngeneic mouse mammary tumor model, we found that breast tumor cells derived from TDLN have higher malignancy and removal of TDLNs significantly reduced distant metastasis. Up-regulation of oncogenic Il-17rb in cancer cells derived from TDLNs contributes to their malignancy. TGF-ß1 secreted from regulatory T cells (Tregs) in the TDLNs mediated the up-regulation of Il-17rb through downstream Smad2/3/4 signaling. These phenotypes can be abolished by TGF-ß1 neutralization or depletion of Tregs. Consistently, clinical data showed that the up-regulation of IL-17RB in cancer cells from LN metastases correlated with the increased prevalence of Tregs as well as the aggressive growth of tumors in mouse xenograft assay. Together, these results indicate that Tregs in TDLNs play an important role in modulating the malignancy of breast cancer cells for distant metastasis. Blocking IL-17RB expression could therefore be a potential approach to curb the process.


Subject(s)
Breast Neoplasms/pathology , Lymph Nodes/pathology , Receptors, Interleukin-17/metabolism , T-Lymphocytes, Regulatory/metabolism , Transforming Growth Factor beta1/metabolism , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/radiotherapy , Female , Humans , Lymph Nodes/immunology , Lymphatic Metastasis , Mammary Neoplasms, Animal/metabolism , Mammary Neoplasms, Animal/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred NOD , Mice, SCID , RNA, Small Interfering/metabolism , Receptors, Interleukin-17/antagonists & inhibitors , Receptors, Interleukin-17/genetics , Signal Transduction , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , Transplantation, Homologous , Tumor Cells, Cultured , Up-Regulation
10.
Proc Natl Acad Sci U S A ; 113(4): 960-5, 2016 Jan 26.
Article in English | MEDLINE | ID: mdl-26677875

ABSTRACT

The discovery of cancer stem cells (CSCs), which are responsible for self-renewal and tumor growth in heterogeneous cancer tissues, has stimulated interests in developing new cancer therapies and early diagnosis. However, the markers currently used for isolation of CSCs are often not selective enough to enrich CSCs for the study of this special cell population. Here we show that the breast CSCs isolated with CD44(+)CD24(-/lo)SSEA-3(+) or ESA(hi)PROCR(hi)SSEA-3(+) markers had higher tumorigenicity than those with conventional markers in vitro and in vivo. As few as 10 cells with CD44(+)CD24(-/lo)SSEA-3(+) formed tumor in mice, compared with more than 100 cells with CD44(+)CD24(-/lo). Suppression of SSEA-3 expression by knockdown of the gene encoding ß-1,3-galactosyltransferase 5 (ß3GalT5) in the globo-series pathway, led to apoptosis in cancer cells specifically but had no effect on normal cells. This finding is further supported by the analysis of SSEA-3 and the two related globo-series epitopes SSEA4 and globo-H in stem cells (embryonic stem cells and induced pluripotent stem cells) and various normal and cancer cells, and by the antibody approach to target the globo-series glycans and the late-stage clinical trials of a breast cancer vaccine.


Subject(s)
Antigens, Tumor-Associated, Carbohydrate/analysis , Biomarkers, Tumor/analysis , Breast Neoplasms/pathology , Galactosyltransferases/analysis , Neoplastic Stem Cells/chemistry , Stage-Specific Embryonic Antigens/analysis , Animals , Apoptosis , Base Sequence , Cell Line, Tumor , Female , Humans , Mice , Molecular Sequence Data
11.
J Exp Med ; 212(3): 333-49, 2015 Mar 09.
Article in English | MEDLINE | ID: mdl-25732306

ABSTRACT

Pancreatic cancer has an extremely high mortality rate due to its aggressive metastatic nature. Resolving the underlying mechanisms will be crucial for treatment. Here, we found that overexpression of IL-17B receptor (IL-17RB) strongly correlated with postoperative metastasis and inversely correlated with progression-free survival in pancreatic cancer patients. Consistently, results from ex vivo experiments further validated that IL-17RB and its ligand, IL-17B, plays an essential role in pancreatic cancer metastasis and malignancy. Signals from IL-17B-IL-17RB activated CCL20/CXCL1/IL-8/TFF1 chemokine expressions via the ERK1/2 pathway to promote cancer cell invasion, macrophage and endothelial cell recruitment at primary sites, and cancer cell survival at distant organs. Treatment with a newly derived monoclonal antibody against IL-17RB blocked tumor metastasis and promoted survival in a mouse xenograft model. These findings not only illustrate a key mechanism underlying the highly aggressive characteristics of pancreatic cancer but also provide a practical approach to tackle this disease.


Subject(s)
Antibodies, Monoclonal/pharmacology , Chemokines/metabolism , Interleukin-17/immunology , Interleukin-17/metabolism , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Receptors, Interleukin-17/metabolism , Animals , Disease-Free Survival , Female , Gene Expression Regulation, Neoplastic , Humans , Interleukin-17/genetics , Male , Mice, SCID , Middle Aged , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/mortality , Receptors, Interleukin-17/genetics , Signal Transduction , Xenograft Model Antitumor Assays
12.
Proc Natl Acad Sci U S A ; 110(30): 12331-6, 2013 Jul 23.
Article in English | MEDLINE | ID: mdl-23836662

ABSTRACT

The circadian clock gene Period2 (PER2) has been suggested to be a tumor suppressor. However, detailed mechanistic evidence has not been provided to support this hypothesis. We found that loss of PER2 enhanced invasion and activated expression of epithelial-mesenchymal transition (EMT) genes including TWIST1, SLUG, and SNAIL. This finding was corroborated by clinical observation that PER2 down-regulation was associated with poor prognosis in breast cancer patients. We further demonstrated that PER2 served as a transcriptional corepressor, which recruited polycomb proteins EZH2 and SUZ12 as well as HDAC2 to octamer transcription factor 1 (OCT1) (POU2F1) binding sites of the TWIST1 and SLUG promoters to repress expression of these EMT genes. Hypoxia, a condition commonly observed in tumors, caused PER2 degradation and disrupted the PER2 repressor complex, leading to activation of EMT gene expression. This result was further supported by clinical data showing a significant negative correlation between hypoxia and PER2. Thus, our findings clearly demonstrate the tumor suppression function of PER2 and elucidate a pathway by which hypoxia promotes EMT via degradation of PER2.


Subject(s)
Breast Neoplasms/metabolism , Epithelial-Mesenchymal Transition , Gene Expression Regulation/genetics , Hypoxia/genetics , Organic Cation Transporter 1/physiology , Period Circadian Proteins/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Down-Regulation , Female , Humans , Promoter Regions, Genetic , Protein Processing, Post-Translational , Up-Regulation/genetics
13.
Cancer Res ; 72(18): 4652-61, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-22826604

ABSTRACT

Tumor microenvironment plays a critical role in regulating tumor progression by secreting factors that mediate cancer cell growth. Stromal fibroblasts can promote tumor growth through paracrine factors; however, restraint of malignant carcinoma progression by the microenvironment also has been observed. The mechanisms that underlie this paradox remain unknown. Here, we report that the tumorigenic potential of breast cancer cells is determined by an interaction between the Robo1 receptor and its ligand Slit2, which is secreted by stromal fibroblasts. The presence of an active Slit2/Robo1 signal blocks the translocation of ß-catenin into nucleus, leading to downregulation of c-myc and cyclin D1 via the phosphoinositide 3-kinase (PI3K)/Akt pathway. Clinically, high Robo1 expression in the breast cancer cells correlates with increased survival in patients with breast cancer, and low Slit2 expression in the stromal fibroblasts is associated with lymph node metastasis. Together, our findings explain how a specific tumor microenvironment can restrain a given type of cancer cell from progression and show that both stromal fibroblasts and tumor cell heterogeneity affect breast cancer outcomes.


Subject(s)
Breast Neoplasms/metabolism , Fibroblasts/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Nerve Tissue Proteins/metabolism , Receptors, Immunologic/metabolism , Signal Transduction/physiology , Tumor Microenvironment/physiology , Animals , Cell Line, Tumor , Disease Progression , Female , Humans , Immunoblotting , Immunohistochemistry , Immunoprecipitation , Mice , Mice, Inbred NOD , Mice, SCID , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Real-Time Polymerase Chain Reaction , beta Catenin/metabolism , Roundabout Proteins
14.
J Mol Biomark Diagn ; Suppl 8: 6, 2012.
Article in English | MEDLINE | ID: mdl-24977105

ABSTRACT

Breast cancer is the most common cancer and the second leading cause of death in U.S. women. Due to early detection and advanced treatment, the breast cancer death rate has been declining since 1990. However, disease recurrence is still the major obstacle in moving from therapy to truly curative treatments. Recent evidence has indicated that breast cancer recurrence is often caused by a subpopulation of breast cancer cells. This subset of cancer cells, usually referred to as breast cancer stem cells (BCSCs), exhibits stem cell phenotypes. They can self-renew and asymmetrically divide to more differentiated cancer cells. These cells are also highly resistant to conventional therapeutic reagents. Therefore, identifying and characterizing these BCSC subpopulations within the larger population of breast cancer cells is essential for developing new strategies to treat breast cancer and prevent recurrence. In this review article, we discuss the current proposed model for the origin of tumor heterogeneity, summarize the recent findings of cell surface and cytoplasmic markers for BCSC identification, review the regulatory mechanisms by which BCSCs maintain or non-cancer stem cells acquire BCSC characteristics, describe the proposed strategies to eliminate BCSCs, and highlight the current limitations and challenges to translate basic BCSC research to clinical application including establishment of clinical biomarkers and therapeutic treatments specifically targeting BCSCs.

15.
Curr Opin Pharmacol ; 10(6): 698-705, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20833107

ABSTRACT

Hepatocyte nuclear factor 4α (HNF4α) is a highly conserved member of the nuclear receptor superfamily of ligand-dependent transcription factors. It is best known as a master regulator of liver-specific gene expression, especially those genes involved in lipid transport and glucose metabolism. However, there is also a growing body of work that indicates the importance of HNF4α in the regulation of genes involved in xenobiotic and drug metabolism. A recent study identifying the essential fatty acid linoleic acid (LA, C18:2) as the endogenous, reversible ligand for HNF4α suggests that HNF4α may also be a potential drug target and that its activity may be regulated by diet. This review will discuss the role of HNF4α in drug metabolism, including the genes it regulates, the factors that regulate its activity, and its potential as a drug target.


Subject(s)
Gene Expression Regulation , Hepatocyte Nuclear Factor 4/metabolism , Inactivation, Metabolic , Pharmaceutical Preparations/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Cytochrome P-450 Enzyme System/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Female , Gene Expression , Hepatocyte Nuclear Factor 4/genetics , Humans , Liver/metabolism , Male , Molecular Targeted Therapy , Regulatory Elements, Transcriptional , Sex Characteristics , Transcription Factors/genetics , Transcription Factors/metabolism
16.
PLoS One ; 4(12): e8377, 2009 Dec 21.
Article in English | MEDLINE | ID: mdl-20027313

ABSTRACT

Heterogeneity of cancer stem/progenitor cells that give rise to different forms of cancer has been well demonstrated for leukemia. However, this fundamental concept has yet to be established for solid tumors including breast cancer. In this communication, we analyzed solid tumor cancer stem cell markers in human breast cancer cell lines and primary specimens using flow cytometry. The stem/progenitor cell properties of different marker expressing-cell populations were further assessed by in vitro soft agar colony formation assay and the ability to form tumors in NOD/SCID mice. We found that the expression of stem cell markers varied greatly among breast cancer cell lines. In MDA-MB-231 cells, PROCR and ESA, instead of the widely used breast cancer stem cell markers CD44(+)/CD24(-/low) and ALDH, could be used to highly enrich cancer stem/progenitor cell populations which exhibited the ability to self renew and divide asymmetrically. Furthermore, the PROCR(+)/ESA(+) cells expressed epithelial-mesenchymal transition markers. PROCR could also be used to enrich cells with colony forming ability from MB-361 cells. Moreover, consistent with the marker profiling using cell lines, the expression of stem cell markers differed greatly among primary tumors. There was an association between metastasis status and a high prevalence of certain markers including CD44(+)/CD24(-/low), ESA(+), CD133(+), CXCR4(+) and PROCR(+) in primary tumor cells. Taken together, these results suggest that similar to leukemia, several stem/progenitor cell-like subpopulations can exist in breast cancer.


Subject(s)
Biomarkers, Tumor/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Lineage , Flow Cytometry/methods , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Agar , Animals , Antigens, CD/metabolism , CD24 Antigen/metabolism , Cell Division , Cell Line, Tumor , Cell Separation , Endothelial Protein C Receptor , Epithelium/metabolism , Epithelium/pathology , Female , Humans , Hyaluronan Receptors/metabolism , Mesoderm/metabolism , Mesoderm/pathology , Mice , Mice, SCID , Receptors, Cell Surface/metabolism , Tumor Stem Cell Assay , Xenograft Model Antitumor Assays
17.
J Formos Med Assoc ; 107(10): 751-66, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18926942

ABSTRACT

Studies of breast cancer stem cells are in their infancy and many fundamental questions have yet to be fully addressed. The molecular distinction between normal and cancerous breast stem cells is not clear. While there have been recent breakthroughs in mouse mammary stem cells and lineage determination in mammary glands, little has been determined in human cells. Microarray analyses have provided molecular categorization of breast cancer. However, the cellular origin of different types of breast cancer is largely unknown. In addition, the relationship between breast cancer stem cells and mammary progenitor cells has yet to be clarified. One of the key questions is how a normal mammary stem cell becomes a breast cancer stem cell. Importantly, the existence of different types of human breast cancers with distinct pathologic and molecular signatures suggests the possibility that different types of breast cancer stem cells may exist. Here, we aim to review the current evidence for the existence of different subtypes of breast cancer stem cells and provide further insight into how tumor suppressors might be involved in the initiation of breast cancer stem cells.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/pathology , Genes, Tumor Suppressor/physiology , Neoplastic Stem Cells/physiology , Animals , Breast Neoplasms/therapy , Female , Humans , Mice
18.
Mol Endocrinol ; 22(1): 78-90, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17885207

ABSTRACT

The dichotomy between cellular differentiation and proliferation is a fundamental aspect of both normal development and tumor progression; however, the molecular basis of this opposition is not well understood. To address this issue, we investigated the mechanism by which the nuclear receptor hepatocyte nuclear factor 4alpha1 (HNF4alpha1) regulates the expression of the human cyclin-dependent kinase inhibitor gene p21/WAF1 (CDKN1A). We found that HNF4alpha1, a transcription factor that plays a central role in differentiation in the liver, pancreas, and intestine, activates the expression of p21 primarily by interacting with promoter-bound Sp1 at both the proximal promoter region and at newly identified sites in a distal region (-2.4 kb). Although HNF4alpha1 also binds two additional regions containing putative HNF4alpha binding sites, HNF4alpha1 mutants deficient in DNA binding activate the p21 promoter to the same extent as wild-type HNF4alpha1, indicating that direct DNA binding by HNF4alpha1 is not necessary for p21 activation. We also observed an in vitro and in vivo interaction between HNF4alpha1 and c-Myc as well as a competition between these two transcription factors for interaction with promoter-bound Sp1 and regulation of p21. Finally, we show that c-Myc competes with HNF4alpha1 for control of apolipoprotein C3 (APOC3), a gene associated with the differentiated hepatic phenotype. These results suggest a general model by which a differentiation factor (HNF4alpha1) and a proliferation factor (c-Myc) may compete for control of genes involved in cell proliferation and differentiation.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p21/genetics , Hepatocyte Nuclear Factor 4/metabolism , Promoter Regions, Genetic/genetics , Proto-Oncogene Proteins c-myc/metabolism , Adenoviridae/genetics , Binding Sites/genetics , Cell Line, Tumor , Cell Proliferation , Electrophoresis, Polyacrylamide Gel , Hepatocyte Nuclear Factor 4/genetics , Humans , Immunoprecipitation , Oligonucleotide Array Sequence Analysis , Protein Binding , Proto-Oncogene Proteins c-myc/genetics , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Sp1 Transcription Factor/genetics , Sp1 Transcription Factor/metabolism , Transfection , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
19.
Biochem J ; 400(2): 303-13, 2006 Dec 01.
Article in English | MEDLINE | ID: mdl-16895524

ABSTRACT

The liver is exposed to a wide variety of toxic agents, many of which damage DNA and result in increased levels of the tumour suppressor protein p53. We have previously shown that p53 inhibits the transactivation function of HNF (hepatocyte nuclear factor) 4alpha1, a nuclear receptor known to be critical for early development and liver differentiation. In the present study we demonstrate that p53 also down-regulates expression of the human HNF4alpha gene via the proximal P1 promoter. Overexpression of wild-type p53 down-regulated endogenous levels of both HNF4alpha protein and mRNA in Hep3B cells. This decrease was also observed when HepG2 cells were exposed to UV irradiation or doxorubicin, both of which increased endogenous p53 protein levels. Ectopically expressed p53, but not a mutant p53 defective in DNA binding (R249S), down-regulated HNF4alpha P1 promoter activity. Chromatin immunoprecipitation also showed that endogenous p53 bound the HNF4alpha P1 promoter in vivo after doxorubicin treatment. The mechanism by which p53 down-regulates the P1 promoter appears to be multifaceted. The down-regulation was partially recovered by inhibition of HDAC activity and appears to involve the positive regulator HNF6alpha. p53 bound HNF6alpha in vivo and in vitro and prevented HNF6alpha from binding DNA in vitro. p53 also repressed stimulation of the P1 promoter by HNF6alpha in vivo. However, since the R249S p53 mutant also bound HNF6alpha, binding HNF6alpha is apparently not sufficient for the repression. Implications of the p53-mediated repression of HNF4alpha expression in response to cellular stress are discussed.


Subject(s)
Carcinoma, Hepatocellular/genetics , Hepatocyte Nuclear Factor 4/genetics , Liver Neoplasms/genetics , Tumor Suppressor Protein p53/genetics , Adenoviridae/genetics , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Down-Regulation/drug effects , Down-Regulation/radiation effects , Doxorubicin/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/radiation effects , Hepatocyte Nuclear Factor 4/biosynthesis , Hepatocyte Nuclear Factor 6/antagonists & inhibitors , Hepatocyte Nuclear Factor 6/genetics , Histone Deacetylases/metabolism , Humans , Liver Neoplasms/metabolism , Promoter Regions, Genetic , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Transcriptional Activation , Transfection , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/radiation effects , Ultraviolet Rays
SELECTION OF CITATIONS
SEARCH DETAIL
...