Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Article in English | MEDLINE | ID: mdl-28785242

ABSTRACT

Activating transcription factor-5 (ATF5) is an anti-apoptotic factor and has been implicated in enhancing the survival of cancer cells under stress and in regulating the autophagy process. Targeting ATF5 in anticancer therapy may be particularly attractive because of its differential role in cancer cells than in non-transformed cells, thus allowing specificity of the treatment. Using the delivery of short hairpin RNA vectors into the Mvt1 and Met1 cell lines, we tested the role of ATF5 in the development of mammary tumors in vivo and in regulating proliferation and migration of these cells in vitro. In this study, we demonstrate that knockdown of ATF5 (ATF5-KD) in both cell lines results in a decreased tumor volume and weight, as well as in a reduced proliferation rate and migratory potential of the cells. In addition, ATF5-KD led to an increased autophagy flux and a shift in the sub-populations comprising Mvt1 cells from the aggressive CD24-positive cells toward less aggressive CD24-negative cells. Taken together, these findings suggest that ATF5 plays an important role in enhancing mammary tumor cells overall aggressiveness and in promoting mammary tumor growth and emphasize the possible benefit of anti-ATF5 therapy in breast cancer patients, particularly, against tumors characterized with the positive expression of cell surface CD24.

2.
Breast Cancer Res ; 18(1): 51, 2016 05 14.
Article in English | MEDLINE | ID: mdl-27179633

ABSTRACT

BACKGROUND: The pro-tumorigenic effects of the insulin-like growth factor receptor (IGF1R) are well described. IGF1R promotes cancer cell survival and proliferation and prevents apoptosis, and, additionally it was shown that IGF1R levels are significantly elevated in most common human malignancies including breast cancer. However, results from phase 3 clinical trials in unselected patients demonstrated lack of efficacy for anti-IGF1R therapy. These findings suggest that predictive biomarkers are greatly warranted in order to identify patients that will benefit from anti-IGF1R therapeutic strategies. METHODS: Using the delivery of shRNA vectors into the Mvt1 cell line, we tested the role of the IGF1R in the development of mammary tumors. Based on CD24 cell surface expression, control and IGF1R-knockdown (IGF1R-KD) cells were FACS sorted into CD24(-) and CD24(+) subsets and further characterized in vitro. The tumorigenic capacity of each was determined following orthotopic inoculation into the mammary fat pad of female mice. Tumor cells were FACS characterized upon sacrifice to determine IGF1R effect on the plasticity of this cell's phenotype. Metastatic capacity of the cells was assessed using the tail vein assay. RESULTS: In this study we demonstrate that downregulation of the IGF1R specifically in cancer cells expressing CD24 on the cell surface membrane affect both their morphology (from mesenchymal-like into epithelial-like morphology) and phenotype in vitro. Moreover, we demonstrate that IGF1R-KD abolished both CD24(+) cells capacity to form mammary tumors and lung metastatic lesions. We found in both cells and tumors a marked upregulation in CTFG and a significant reduction of SLP1 expression in the CD24(+)/IGF1R-KD; tumor-suppressor and tumor-promoting genes respectively. Moreover, we demonstrate here that the IGF1R is essential for the maintenance of stem/progenitor-like cancer cells and we further demonstrate that IGF1R-KD induces in vivo differentiation of the CD24(+) cells toward the CD24(-) phenotype. This further supports the antitumorigenic effects of IGF1R-KD, as we recently published that these differentiated cells demonstrate significantly lower tumorigenic capacity compared with their CD24(+) counterparts. CONCLUSIONS: Taken together these findings suggest that CD24 cell surface expression may serve as a valuable biomarker in order to identify mammary tumors that will positively respond to targeted IGF1R therapies.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/metabolism , CD24 Antigen/metabolism , Gene Expression , Receptor, IGF Type 1/genetics , Animals , Biomarkers , Breast Neoplasms/pathology , CD24 Antigen/genetics , Cell Line, Tumor , Connective Tissue Growth Factor/genetics , Connective Tissue Growth Factor/metabolism , Disease Models, Animal , Female , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Immunophenotyping , Mammary Neoplasms, Animal , Mice , Neoplasm Metastasis , RNA Interference , Receptor, IGF Type 1/metabolism , Secretory Leukocyte Peptidase Inhibitor/genetics , Secretory Leukocyte Peptidase Inhibitor/metabolism
3.
Handb Exp Pharmacol ; 233: 355-72, 2016.
Article in English | MEDLINE | ID: mdl-25903410

ABSTRACT

Obesity is associated with multiple metabolic disorders that drive cardiovascular disease, T2D and cancer. The doubling in the number of obese adults over the past 3 decades led to the recognition of obesity as a "disease". With over 42 million children obese or overweight, this epidemic is rapidly growing worldwide. Obesity and T2D are both associated together and independently with an increased risk for cancer and a worse prognosis. Accumulating evidence from epidemiological studies revealed potential factors that may explain the association between obesity-linked metabolic disorders and cancer risk. Studies based on the insulin resistance MKR mice, highlighted the roe of the insulin receptor and its downstream signaling proteins in mediating hyperinsulinemia's mitogenic effects. Hypercholesterolemia was also shown to promote the formation of larger tumors and enhancement in metastasis. Furthermore, the conversion of cholesterol into 27-Hydroxycholesterol was found to link high fat diet-induced hypercholesterolemia with cancer pathophysiology. Alteration in circulating adipokines and cytokines are commonly found in obesity and T2D. Adipokines are involved in tumor growth through multiple mechanisms including mTOR, VEGF and cyclins. In addition, adipose tissues are known to recruit and alter macrophage phenotype; these macrophages can promote cancer progression by secreting inflammatory cytokines such as TNF-α and IL-6. Better characterization on the above factors and their downstream effects is required in order to translate the current knowledge into the clinic, but more importantly is to understand which are the key factors that drive cancer in each patient. Until we reach this point, policies and activities toward healthy diets and physical activities remain the best medicine.


Subject(s)
Diabetes Mellitus, Type 2/complications , Metabolic Syndrome/complications , Neoplasms/etiology , Adipokines/physiology , Animals , Cytokines/physiology , Diabetes Mellitus, Type 2/epidemiology , Disease Models, Animal , Humans , Obesity/complications , Obesity/epidemiology
4.
J Endocrinol ; 227(3): 143-151, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26383532

ABSTRACT

Obesity and type 2 diabetes (T2D) are associated with an increased risk of breast cancer incidence and mortality. Common features of obesity and T2D are insulin resistance and hyperinsulinemia. A mammary tumor promoting effect of insulin resistance and hyperinsulinemia was demonstrated in the transgenic female MKR mouse model of pre-diabetes inoculated with mammary cancer cells. Interestingly, in MKR mice, as well as in other diabetic mouse models, males exhibit severe hyperglycemia, while females display insulin resistance and hyperinsulinemia with only a mild increase in blood glucose levels. This gender-specific protection from hyperglycemia may be attributed to estradiol, a key player in the regulation of the metabolic state, including obesity, glucose homeostasis, insulin resistance, and lipid profile. The aim of this study was to investigate the effects of ovariectomy (including the removal of endogenous estradiol) on the metabolic state of MKR female mice and subsequently on the growth of Mvt-1 mammary cancer cells, inoculated into the mammary fat pad of ovariectomized mice, compared with sham-operated mice. The results showed an increase in body weight, accompanied by increased fat mass, elevated blood glucose levels, and hypercholesterolemia, in ovariectomized MKR mice. In addition, mammary tumor growth was significantly higher in these mice. The results suggest that ovarian hormone deficiency may promote impaired metabolic homeostasis in the hyperinsulinemic MKR female mice, which in turn is associated with an increased growth of mammary tumors.


Subject(s)
Mammary Neoplasms, Experimental/etiology , Mammary Neoplasms, Experimental/pathology , Ovariectomy/adverse effects , Prediabetic State/pathology , Animals , Blood Glucose/metabolism , Female , Insulin Resistance , Mice , Mice, Transgenic , Neoplasm Transplantation , Prediabetic State/genetics
5.
Breast Cancer Res ; 17: 78, 2015 Jun 04.
Article in English | MEDLINE | ID: mdl-26040280

ABSTRACT

INTRODUCTION: Breast tumors are comprised of distinct cancer cell populations which differ in their tumorigenic and metastatic capacity. Characterization of cell surface markers enables investigators to distinguish between cancer stem cells and their counterparts. CD24 is a well-known cell surface marker for mammary epithelial cells isolation, recently it was suggested as a potential prognostic marker in a wide variety of malignancies. Here, we demonstrate that CD24(+) cells create intra-tumor heterogeneity, and display highly metastatic properties. METHODS: The mammary carcinoma Mvt1 cells were sorted into CD24(-) and CD24(+) cells. Both subsets were morphologically and phenotypically characterized, and tumorigenic capacity was assessed via orthotopic inoculation of each subset into the mammary fat pad of wild-type and MKR mice. The metastatic capacity of each subset was determined with the tail vein metastasis assay. The role of CD24 in tumorigenesis was further examined with shRNA technology. GFP-labeled cells were monitored in vivo for differentiation. The genetic profile of each subset was analyzed using RNA sequencing. RESULTS: CD24(+) cells displayed a more spindle-like cytoplasm. The cells formed mammospheres in high efficiency and CD24(+) tumors displayed rapid growth in both WT and MKR mice, and were more metastatic than CD24- cells. Interestingly, CD24-KD in CD24+ cells had no effect both in vitro and in vivo on the various parameters studied. Moreover, CD24(+) cells gave rise in vivo to the CD24(-) that comprised the bulk of the tumor. RNA-seq analysis revealed enrichment of genes and pathways of the extracellular matrix in the CD24(+) cells. CONCLUSION: CD24(+) cells account for heterogeneity in mammary tumors. CD24 expression at early stages of the cancer process is an indication of a highly invasive tumor. However, CD24 is not a suitable therapeutic target; instead we suggest here new potential targets accounting for early differentiated cancer cells tumorigenic capacity.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , CD24 Antigen/metabolism , Animals , Biomarkers , Breast Neoplasms/genetics , CD24 Antigen/genetics , Cell Line, Tumor , Cell Transformation, Neoplastic/metabolism , Disease Models, Animal , Extracellular Matrix/genetics , Extracellular Matrix/metabolism , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Gene Knockout Techniques , Immunophenotyping , Mice , Neoplasm Metastasis , Neoplastic Stem Cells/metabolism , Phenotype , Tumor Burden
6.
Endocr Relat Cancer ; 22(2): 145-57, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25694511

ABSTRACT

Accumulating evidence from clinical trials indicates that specific targeting of the IGF1 receptor (IGF1R) is not efficient as an anti-breast cancer treatment. One possible reason is that the mitogenic signals from the insulin receptor (IR) can be processed independently or as compensation to inhibition of the IGF1R. In this study, we highlight the role of the IR in mediating breast tumor progression in both WT mice and a hyperinsulinemic MKR mouse model by induction of Ir (Insr) or Igf1r knockdown (KD) in the mammary carcinoma Mvt-1 cell line. By using the specific IR antagonist-S961, we demonstrated that Igf1r-KD induces elevated responses by the IR to IGF1. On the other hand, Ir-KD cells generated significantly smaller tumors in the mammary fat pads of both WT and MKR mice, as opposed to control cells, whereas the Igf1r-KD cells did not. The tumorigenic effects of insulin on the Mvt-1 cells were also demonstrated using microarray analysis, which indicates alteration of genes and signaling pathways involved in proliferation, the cell cycle, and apoptosis following insulin stimulation. In addition, the correlation between IR and the potential prognostic marker for aggressive breast cancer, CD24, was examined in the Ir-KD cells. Fluorescence-activated cell sorting (FACS) analysis revealed more than 60% reduction in CD24 expression in the Ir-KD cells when compared with the control cells. Our results also indicate that CD24-expressing cells can restore, at least in part, the tumorigenic capacity of Ir-KD cells. Taken together, our results highlight the mitogenic role of the IR in mammary tumor progression with a direct link to CD24 expression.


Subject(s)
Mammary Neoplasms, Animal/metabolism , Receptor, Insulin/metabolism , Animals , CD24 Antigen/metabolism , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Insulin/pharmacology , Mammary Neoplasms, Animal/pathology , Mice , Mice, Transgenic , Mitogen-Activated Protein Kinases/metabolism , Oligonucleotide Array Sequence Analysis , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptor, IGF Type 1/metabolism , Signal Transduction , Tumor Burden
7.
Peptides ; 40: 133-40, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23340021

ABSTRACT

Bradykinin (BK) is a major potent inflammatory mediator outside the central nervous system. In Alzheimer's disease, BK release and BK receptor expression in brain tissues are upregulated relatively early during the course of the disease. Hence, BK was believed to promote neuroinflammation. However, BK was recently reported to possess anti-inflammatory and neuroprotective roles. Exposure of BV2 microglial cell line to BK lead to a decrease in NO release from unstimulated cells as well as a dose-dependent attenuation, mediated by both B1 and B2 receptors, in lipopolysaccharide (LPS)-induced NO production. In this study we examined whether cyclic adenosine monophosphate (cAMP) signaling is involved in BK-mediated effect in microglial nitric oxide (NO) production. A protein kinase A (PKA) inhibitor mimicked the effects of BK, while cAMP elevating agents antagonized BK-mediated NO decrease. Moreover, BK inhibited the activation of cAMP responsive element binding protein (CREB). In addition, BK protected microglial cells from death triggered by combinations of LPS and each of the cAMP elevating agents. Finally, the addition of Gαi protein inhibitor abrogated the effects of BK on NO release, and the expression of Gαi protein in the plasma membrane was induced by BK. These results suggest that BK-mediated reduction in microglial NO production depends on coupling to Gi protein and also involves inhibition of cAMP-PKA-CREB signaling.


Subject(s)
Alzheimer Disease/metabolism , Bradykinin/administration & dosage , Cyclic AMP/metabolism , Nitric Oxide/metabolism , Signal Transduction/drug effects , Alzheimer Disease/complications , Alzheimer Disease/pathology , Animals , Bradykinin/metabolism , Cell Line , Cyclic AMP Response Element-Binding Protein/metabolism , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Humans , Inflammation/metabolism , Inflammation/pathology , Lipopolysaccharides/pharmacology , Mice , Microglia/cytology , Microglia/metabolism , Neurons/drug effects , Neurons/pathology , Receptors, Bradykinin/metabolism
8.
J Neuroinflammation ; 7: 61, 2010 Oct 11.
Article in English | MEDLINE | ID: mdl-20937084

ABSTRACT

Microglial activation plays an integral role in the development and course of neurodegeneration. Although neuropeptides such as bradykinin (BK), somatostatin (SST), and endothelin (ET) are known to be important mediators of inflammation in the periphery, evidence of a similar function in brain is scarce. Using immunocytochemistry, we demonstrate the expression of receptors for BK (B1, B2 subtypes), ET (ETA, ETB subtypes) and SST (SST 2, 3, 4 subtypes) in primary microglia and microglial cell lines. Exposure of BV2 and N9, as well as primary microglial cells to BK or SST increased Aß uptake in a concentration-dependent manner, whereas endothelin decreased Aß uptake. This was caused by increased phagocytosis of Aß since the rate of intracellular Aß degradation remained unaffected. All neuropeptides increased chemotactic activity of microglia. In addition, BK reduced Aß-induced expression of proinflammatory genes including iNOS and COX-2. ET decreased the Aß-induced expression of monocyte chemoattractant protein 1 and interleukin-6. These results suggest that neuropeptides play an important role in chemotaxis and Aß clearance and modulate the brain's response to neuroinflammatory processes.


Subject(s)
Amyloid beta-Peptides/metabolism , Cell Movement/physiology , Microglia/metabolism , Phagocytosis/physiology , Receptors, Bradykinin/metabolism , Receptors, Endothelin/metabolism , Receptors, Somatostatin/metabolism , Analysis of Variance , Animals , Blotting, Western , Cell Line , Cells, Cultured , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Immunohistochemistry , Mice , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Reverse Transcriptase Polymerase Chain Reaction
9.
Regul Pept ; 150(1-3): 21-5, 2008 Oct 09.
Article in English | MEDLINE | ID: mdl-18619496

ABSTRACT

Glial inflammation plays an integral role in the development of neurodegenerative disease. Although somatostatin is known to be a local anti-inflammatory factor in the periphery, evidence of a similar function in the brain is scarce. The aim of the present study was to investigate the effect of somatostatin on prostaglandin E(2) synthesis in primary neonatal rat glial cells. The data shows that high concentrations of somatostatin (10(-5)-10(-4)) significantly increased prostaglandin synthesis. By contrast, when used at physiologically relevant concentrations (10(-9)-10(-7) M), somatostatin and somatostatin receptor agonists decreased prostaglandin E(2) synthesis in non-stimulated glial cells as well as in lipopolysaccharide-induced prostaglandin synthesis. The inhibitory effect of somatostatin in lipopolysaccharide-treated cells could be mimicked by protein kinase A inhibitor and was prevented by forskolin. These observations suggest the presence of a novel neuro-immune feedback pathway through which somatostatin inhibits glial prostaglandin synthesis, and thus may prove to play a role in brain inflammation. This action of somatostatin may have a therapeutic potential in pathological conditions of the brain, where an inflammatory response is involved.


Subject(s)
Dinoprostone/biosynthesis , Encephalitis/metabolism , Hormone Antagonists/metabolism , Neuroglia/metabolism , Somatostatin/metabolism , Amides/pharmacology , Animals , Animals, Newborn , Brain/cytology , Cells, Cultured , Colforsin/pharmacology , Culture Media/analysis , Culture Media/chemistry , Culture Media, Serum-Free , Dinoprostone/analysis , Dose-Response Relationship, Drug , Hormone Antagonists/pharmacology , Indoles/pharmacology , Isoquinolines/pharmacology , Lipopolysaccharides/pharmacology , Neuroglia/drug effects , Nitrobenzenes/pharmacology , Octreotide/pharmacology , Radioimmunoassay , Rats , Rats, Wistar , Receptors, Somatostatin/metabolism , Somatostatin/analogs & derivatives , Somatostatin/pharmacology , Sulfonamides/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...