Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Cells ; 10(6)2021 06 15.
Article in English | MEDLINE | ID: mdl-34203746

ABSTRACT

Tremendous data have been accumulated in the effort to understand chemoresistance of triple negative breast cancer (TNBC). However, modifications in cancer cells surviving combined and sequential treatment still remain poorly described. In order to mimic clinical neoadjuvant treatment, we first treated MDA-MB-231 and SUM159-PT TNBC cell lines with epirubicin and cyclophosphamide for 2 days, and then with paclitaxel for another 2 days. After 4 days of recovery, persistent cells surviving the treatment were characterized at both cellular and molecular level. Persistent cells exhibited increased growth and were more invasive in vitro and in zebrafish model. Persistent cells were enriched for vimentinhigh sub-population, vimentin knockdown using siRNA approach decreased the invasive and sphere forming capacities as well as Akt phosphorylation in persistent cells, indicating that vimentin is involved in chemotherapeutic treatment-induced enhancement of TNBC aggressiveness. Interestingly, ectopic vimentin overexpression in native cells increased cell invasion and sphere formation as well as Akt phosphorylation. Furthermore, vimentin overexpression alone rendered the native cells resistant to the drugs, while vimentin knockdown rendered them more sensitive to the drugs. Together, our data suggest that vimentin could be considered as a new targetable player in the ever-elusive status of drug resistance and recurrence of TNBC.


Subject(s)
Drug Resistance, Neoplasm/physiology , Triple Negative Breast Neoplasms/metabolism , Vimentin/physiology , Animals , Cell Line, Tumor , Cell Movement/physiology , Cyclophosphamide/pharmacology , Disease Models, Animal , Drug Therapy/methods , Epirubicin/pharmacology , Epithelial-Mesenchymal Transition , Female , Humans , Neoadjuvant Therapy/methods , Neoplasm Invasiveness/pathology , Neoplasm Recurrence, Local , Paclitaxel/therapeutic use , Triple Negative Breast Neoplasms/pathology , Vimentin/metabolism , Zebrafish
2.
Vet Pathol ; 57(4): 507-519, 2020 07.
Article in English | MEDLINE | ID: mdl-32351171

ABSTRACT

Accumulating data highlight the role of neurotrophins and their receptors in human breast cancer. This family includes nerve growth factor (NGF) and brain-derived neurotrophic factor (BDNF), both synthetized as proneurotrophins (proNGF and proBDNF). (pro)NGF and (pro)BDNF initiate their biological effects by binding to both their specific receptors TrkA and TrkB, respectively, and the common receptor p75NTR. Currently, no data are available about their expression and potential role in canine mammary tumors. The aim of this study was to investigate expression of proNGF and BDNF as well as their receptors TrkA, TrkB, and p75NTR in canine mammary carcinomas, and to correlate them with clinicopathological parameters (grade, histological type, lymph node status, recurrence, and distant metastasis) and survival. Immunohistochemistry was performed on serial sections of 96 canine mammary carcinomas with antibodies against proNGF, BDNF, TrkA, TrkB, and p75NTR. Of the 96 carcinomas, proNGF expression was detected in 71 (74%), BDNF in 79 (82%), TrkA in 94 (98%), TrkB in 35 (37%), and p75NTR in 44 (46%). No association was observed between proNGF, BDNF, or TrkA expression and either clinicopathological parameters or survival. TrkB and p75NTR expression were associated with favorable clinicopathological parameters as well as better overall survival.


Subject(s)
Dog Diseases/pathology , Mammary Neoplasms, Animal , Nerve Growth Factors/metabolism , Animals , Brain-Derived Neurotrophic Factor/metabolism , Dogs , Immunohistochemistry/veterinary , Lymph Nodes/pathology , Mammary Neoplasms, Animal/diagnosis , Mammary Neoplasms, Animal/metabolism , Mammary Neoplasms, Animal/pathology , Neoplasm Grading , Neoplasm Metastasis/pathology , Neoplasm Recurrence, Local/veterinary , Nerve Growth Factor/metabolism , Prognosis , Receptor, Nerve Growth Factor/metabolism , Receptor, trkA/metabolism , Receptor, trkB/metabolism
3.
Cancer Lett ; 449: 196-206, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30771434

ABSTRACT

ProNGF expression has been linked to several types of cancers including breast cancer, and we have previously shown that proNGF stimulates breast cancer invasion in an autocrine manner through membrane receptors sortilin and TrkA. However, little is known regarding TrkA-associated protein partners upon proNGF stimulation. By proteomic analysis and proximity ligation assays, we found that proNGF binding to sortilin induced sequential formation of the functional sortilin/TrkA/EphA2 complex, leading to TrkA-phosphorylation dependent Akt activation and EphA2-dependent Src activation. EphA2 inhibition using siRNA approach abolished proNGF-stimulated clonogenic growth of breast cancer cell lines. Combinatorial targeting of TrkA and EphA2 dramatically reduced colony formation in vitro, primary tumor growth and metastatic dissemination towards the brain in vivo. Finally, proximity ligation assay in breast tumor samples revealed that increased TrkA/EphA2 proximity ligation assay signals were correlated with a decrease of overall survival in patients. All together, these data point out the importance of TrkA/EphA2 functional association in proNGF-induced tumor promoting effects, and provide a rationale to target proNGF/TrkA/EphA2 axis by alternative methods other than the simple use of tyrosine kinase inhibitors in breast cancer.


Subject(s)
Breast Neoplasms/enzymology , Cell Proliferation , Ephrin-A2/metabolism , Nerve Growth Factor/metabolism , Protein Precursors/metabolism , Receptor, trkA/metabolism , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Antineoplastic Agents/pharmacology , Brain Neoplasms/enzymology , Brain Neoplasms/prevention & control , Brain Neoplasms/secondary , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Breast Neoplasms/therapy , Ephrin-A2/genetics , Female , Humans , MCF-7 Cells , Mice, SCID , Phosphorylation , Protein Binding , Protein Kinase Inhibitors/pharmacology , RNAi Therapeutics , Receptor, EphA2 , Receptor, trkA/antagonists & inhibitors , Receptor, trkA/genetics , Signal Transduction , Tumor Burden , Xenograft Model Antitumor Assays , src-Family Kinases/metabolism
4.
Vet Pathol ; 56(3): 377-388, 2019 05.
Article in English | MEDLINE | ID: mdl-30558511

ABSTRACT

CD44+/CD24- phenotype has been used to identify human and canine mammary cancer stem-like cells. In canine mammary tumors, CD44+/CD24- phenotype has been associated with high grade and lymph node infiltration. However, several studies have reported opposing results regarding the clinical significance of phenotypic groups formed by the combination of CD44 and CD24 in both human and canine mammary tumors. So far, no study has investigated the correlation between these phenotypes and survival in dogs. The aim of this study was to investigate the expression and distribution of CD44 and CD24 in canine mammary carcinomas and to correlate them with histological diagnosis and survival in a well-characterized cohort. Immunohistochemistry was performed in 96 mammary carcinomas with antibodies against CD44 and CD24. Expression of CD44+ and CD44+/CD24- phenotype was detected in 75 of 96 (78%) and 63 of 96 (65.6%) carcinomas, respectively. Their expression was associated with tumor type, occurring more often in tubular complex carcinomas than in solid carcinomas. CD44+/CD24- phenotype was associated with a better overall survival ( P = .001). CD24+ expression was detected in 52 of 96 tumors (54%) and CD44-/CD24+ phenotype in 39 of 96 tumors (40.6%). Both were associated with poor clinicopathological parameters (high grade, and emboli). No correlation with overall survival was observed. CD44+/CD24- expression was associated with a better prognosis and occurred at high frequency and high level, indicating that this phenotype is not suitable to detect cancer stem cells in canine mammary carcinomas. Although further studies are needed, our results suggest that CD24 may constitute a valuable marker of poor prognosis for canine mammary carcinomas.


Subject(s)
CD24 Antigen/metabolism , Dog Diseases/diagnosis , Hyaluronan Receptors/metabolism , Mammary Neoplasms, Animal/diagnosis , Animals , Dog Diseases/metabolism , Dog Diseases/mortality , Dog Diseases/pathology , Dogs , Female , Mammary Glands, Animal/metabolism , Mammary Glands, Animal/pathology , Mammary Neoplasms, Animal/metabolism , Mammary Neoplasms, Animal/mortality , Mammary Neoplasms, Animal/pathology , Prognosis
5.
Cell Mol Life Sci ; 73(9): 1859-70, 2016 May.
Article in English | MEDLINE | ID: mdl-26883804

ABSTRACT

Cancer stem cells (CSCs), are thought to be at the origin of tumor development and resistance to therapies. Thus, a better understanding of the molecular mechanisms involved in the control of CSC stemness is essential to the design of more effective therapies for cancer patients. Cancer cell stemness and the subsequent expansion of CSCs are regulated by micro-environmental signals including neurotrophins. Over the years, the roles of neurotrophins in tumor development have been well established and regularly reviewed. Especially, nerve growth factor (NGF) and brain-derived neurotrophic factor (BDNF) are reported to stimulate tumor cell proliferation, survival, migration and/or invasion, and favors tumor angiogenesis. More recently, neurotrophins have been reported to regulate CSCs. This review briefly presents neurotrophins and their receptors, summarizes their roles in different cancers, and discusses the emerging evidence of neurotrophins-induced enrichment of CSCs as well as the involved signaling pathways.


Subject(s)
Neoplastic Stem Cells/metabolism , Nerve Growth Factors/metabolism , Animals , Brain-Derived Neurotrophic Factor/metabolism , Epithelial-Mesenchymal Transition , Humans , Neoplastic Stem Cells/cytology , Receptors, Nerve Growth Factor/metabolism , Signal Transduction
6.
J Cell Physiol ; 227(12): 3837-46, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22495877

ABSTRACT

Breast cancer (BC) has a poor prognosis due to its strong metastatic ability. Accumulating data present ether à go-go (hEag1) K(+) channels as relevant player in controlling cell cycle and proliferation of non-invasive BC cells. However, the role of hEag1 in invasive BC cells migration is still unknown. In this study, we studied both the functional expression and the involvement in cell migration of hEag1 in the highly metastatic MDA-MB-231 human BC cells. We showed that hEag1 mRNA and proteins were expressed in human invasive ductal carcinoma tissues and BC cell lines. Functional activity of hEag1 channels in MDA-MB-231 cells was confirmed using astemizole, a hEag1 blocker, or siRNA. Blocking or silencing hEag1 depolarized the membrane potential and reduced both Ca(2+) entry and MDA-MB-231 cell migration without affecting cell proliferation. Recent studies have reported that Ca(2+) entry through Orai1 channels is required for MDA-MB-231 cell migration. Down-regulation of hEag1 or Orai1 reduced Ca(2+) influx and cell migration with similar efficiency. Interestingly, no additive effects on Ca(2+) influx or cell migration were observed in cells co-transfected with sihEag1 and siOrai1. Finally, both Orai1 and hEag1 are expressed in invasive breast adenocarcinoma tissues and invaded metastatic lymph node samples (LNM(+)). In conclusion, this study is the first to demonstrate that hEag1 channels are involved in the serum-induced migration of BC cells by controlling the Ca(2+) entry through Orai1 channels. hEag1 may therefore represent a potential target for the suppression of BC cell migration, and thus prevention of metastasis development.


Subject(s)
Calcium Channels/metabolism , Calcium/metabolism , Cell Movement/physiology , Ether-A-Go-Go Potassium Channels/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Calcium Channels/genetics , Carcinoma, Ductal, Breast/metabolism , Carcinoma, Ductal, Breast/pathology , Cell Line, Tumor , Cell Survival , Ether-A-Go-Go Potassium Channels/genetics , Female , Gene Expression Regulation, Neoplastic/physiology , Humans , Immunohistochemistry , Lymph Nodes/pathology , Manganese , Neoplasm Invasiveness , ORAI1 Protein , Patch-Clamp Techniques , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction
7.
J Membr Biol ; 234(1): 47-56, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20177667

ABSTRACT

Prolactin (PRL) is a polypeptidic hormone which acts both systemically and locally to cause lactation by interacting with the PRL receptor, a Janus kinase (JAK2)-coupled cytokine receptor family member. Several studies have reported that serum PRL level elevation is associated with an increased risk for breast cancer, and evidence has suggested that PRL is one actor in the pathogenesis and progression of this cancer. We previously reported the involvement of hIKCa1 in breast cell cycle progression and cell proliferation. However, mechanisms by which PRL cooperates with these channels to modulate breast epithelial cell proliferation remain unknown. Our results showed that, in the MCF-7 breast cancer cell line, PRL increased hIKCa1 current density. These channels were functional and regulated the resting membrane potential. The PRL effects were inhibited by TRAM-34 and clotrimazole, the most used hIKCa1 blockers. Moreover, PRL increased proliferation in a dose-dependent manner without overexpressing hIKCa1. To determine whether PRL-induced proliferation and hIKCa1 activity involved the JAK2 pathway, we used pharmacological JAK2 inhibitors (AG490 and JAK inhibitor I). Indeed, PRL-induced JAK2 phosphorylation was required for both cell proliferation and hIKCa1 activity. In the presence of either hIKCa1 blockers or siRNA-hIKCa1, PRL failed to increase cell proliferation and hIKCa1 activity. Taken together, our results demonstrate that PRL plays a role in breast cancer cell proliferation by increasing hIKCa1 activity through the JAK2 signaling pathway.


Subject(s)
Intermediate-Conductance Calcium-Activated Potassium Channels/physiology , Janus Kinase 2/physiology , Receptors, Prolactin/physiology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Calcium/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Clotrimazole/pharmacology , Female , Humans , Intermediate-Conductance Calcium-Activated Potassium Channels/drug effects , Prolactin/pharmacology , Pyrazoles , Tyrphostins/pharmacology
8.
Mol Cell Proteomics ; 6(7): 1239-47, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17446556

ABSTRACT

Normal breast epithelial cells are known to exert an apoptotic effect on breast cancer cells, resulting in a potential paracrine inhibition of breast tumor development. In this study we purified and characterized the apoptosis-inducing factors secreted by normal breast epithelial cells. Conditioned medium was concentrated by ultrafiltration and separated on reverse phase Sep-Pak C18 and HPLC. The proapoptotic activity of eluted fractions was tested on MCF-7 breast cancer cells, and nano-LC-nano-ESI-MS/MS allowed the identification of insulin-like growth factor-binding protein-3 (IGFBP-3) and maspin as the proapoptotic factors produced by normal breast epithelial cells. Western blot analysis of conditioned media confirmed the specific secretion of IGFBP-3 and maspin by normal cells but not by breast cancer cells. Immunodepletion of IGFBP-3 and maspin completely abolished the normal cell-induced apoptosis of cancer cells, and recombinant proteins reproduced the effect of normal cell-conditioned medium on apoptosis of breast cancer cells. Together our results indicated that normal breast epithelial cells can induce apoptosis of breast cancer cells through IGFBP-3 and maspin. These findings provide a molecular hypothesis for the long observed inhibitory effect of normal surrounding cells on breast cancer development.


Subject(s)
Apoptosis/physiology , Breast Neoplasms/pathology , Epithelial Cells/physiology , Insulin-Like Growth Factor Binding Protein 3/physiology , Serpins/physiology , Adolescent , Adult , Amino Acid Sequence , Breast Neoplasms/metabolism , Cells, Cultured , Culture Media, Conditioned , Epithelial Cells/metabolism , Female , Humans , Molecular Sequence Data , Proteomics , Tandem Mass Spectrometry
9.
Med Sci (Paris) ; 22(1): 54-61, 2006 Jan.
Article in French | MEDLINE | ID: mdl-16386221

ABSTRACT

Loss of heterozygosity (LOH) of the short arm of chromosome 17 (17p) is one of the most frequent genetic alterations in human cancers. Most often, allelic losses coincide with p53 mutations at 17p13.1. However, in many types of solid tumors including sporadic breast cancers, ovarian cancers, medulloblastomas and small cell lung carcinomas, frequent LOH or DNA methylation changes occur in a more telomeric region at 17p13.3, in absence of any p53 genetic alterations. These results suggest that one or more tumor suppressor genes located at 17p13.3 could be involved in tumorigenesis. In addition, the 17p13.3 region has also been implicated in the Miller-Dieker syndrome (MDS), a severe form of lissencephaly accompanied by developmental anomalies caused by heterozygous gene deletions. Analyses of deletion mapping and CpG island methylation patterns have resulted in the identification of two tumor suppressor genes at 17p13.3, HIC1 (hypermethylated in cancer 1) and OVCA1 (ovarian cancer gene 1). HIC1 is a tumor suppressor gene that encodes a transcriptional repressor with five Krüppel-like C2H2 zinc finger motifs and a N-terminal BTB/POZ domain. Clues to the tumor suppressor function of HIC1 have come from the study of heterozygous Hic1+/- mice, which develop spontaneous malignant tumors of different types. Generation of double heterozygous knockout mice Hic1+/- p53+/- provides strong evidence that epigenetically silenced genes such as HIC1 can significantly influence tumorigenesis driven by mutations of classic tumor suppressor genes. This functional cooperation between HIC1 and p53 is interesting and recently, its has been demonstrated that HIC1 was involved in a certain feedback regulation for p53 in tumor suppression through the histone deacetylase SIRT1. However, despite the fact that epigenetic oncogenesis is one of the most vibrant areas of biologic research, the determinants between genetic versus epigenetic routes of tumor suppressor gene inactivation remain elusive.


Subject(s)
Chromosomes, Human, Pair 17 , DNA-Binding Proteins/genetics , Transcription Factors/genetics , Animals , Chromosome Mapping , Disease Models, Animal , Genetic Markers , Histone Deacetylases/metabolism , Humans , Kruppel-Like Transcription Factors , Loss of Heterozygosity , Mice , Neoplasms/genetics , Tumor Suppressor Protein p53/genetics
10.
Med Sci (Paris) ; 21(4): 405-11, 2005 Apr.
Article in French | MEDLINE | ID: mdl-15811306

ABSTRACT

Epigenetics is defined as "the study of mitotically and/or meiotically heritable changes in gene expression that cannot be explained by changes in the DNA sequence". Setting up the epigenetic program is crucial for correct development and its stable inheritance throughout its lifespan is essential for the maintenance of the tissue- and cell-specific functions of the organism. For many years, the genetic causes of cancer have hold centre stage. However, the recent wealth of information about the molecular mechanisms which, by modulating the chromatin structure, can regulate gene expression has high-lighted the predominant role of epigenetic modifications in the initiation and progression of numerous pathologies, including cancer. The nucleosome is the major target of these epigenetic regulation mechanisms. They include a series of tightly interconnected steps which starting with the setting ("writing") of the epigenetic mark till its "reading" and interpretation will result in long-term gene regulation. The major epigenetic changes associated with tumorigenesis are aberrant DNA methylation of CpG islands located in the promoter region of tumor suppressor gene, global genomic hypomethylation and covalent modifications of histone N-terminal tails which are protruding out from the nucleosome core. In sharp contrast with genetic modifications, epigenetic modifications are highly dynamic and reversible. The characterization of specific inhibitors directed against some key epigenetic players has opened a new and promising therapeutic avenue, the epigenetic therapy, since some inhibitors are already used in clinical trials.


Subject(s)
Epigenesis, Genetic , Neoplasms/genetics , Animals , DNA Methylation , Histones/genetics , Humans
11.
Oncogene ; 23(1): 21-9, 2004 Jan 08.
Article in English | MEDLINE | ID: mdl-14712207

ABSTRACT

Sodium butyrate (NaB) has been proposed as a potential anticancer agent. However, its mechanism of action is not totally elucidated. Here, we showed that NaB-induced cell cycle arrest and apoptosis were associated with an increase of P21(waf1/cip1) in MCF-7 breast cancer cells. This increase was more important in the nuclei, as revealed by immunofluorescence analysis. Transient transfections of MCF-7 cells with p21 deficient for interaction with CDK, but not with p21 deficient for interaction with PCNA (p21PCNA-), abrogated NaB-induced cell cycle arrest. This indicated that cell cycle blockage involved the interaction of P21(waf1/cip1) with CDK. However, P21(waf1/cip1) was dispensable, since p21 antisense did not modify cell cycle arrest. On the other hand, NaB-induced apoptosis was abolished by p21 antisense or p21PCNA-. In addition, NaB decreased PCNA levels, but increased the association of PCNA with P21(waf1/cip1). These results suggested that NaB-induced apoptosis required P21(waf1/cip1) and its interaction with PCNA.


Subject(s)
Apoptosis/drug effects , Butyrates/pharmacology , Cyclins/physiology , G1 Phase/drug effects , Breast Neoplasms/pathology , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p21 , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclins/analysis , DNA Repair , Female , Humans , Plasmids , Proliferating Cell Nuclear Antigen/analysis
12.
Breast Cancer Res Treat ; 76(3): 195-201, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12462380

ABSTRACT

(-)-Epigallocatechin (EGC), one of green tea polyphenols, has been shown to inhibit growth of cancer cells. However its mechanism of action is poorly known. We show here that EGC strongly inhibited the growth of breast cancer cell lines (MCF-7 and MDA-MB-231) but not that of normal breast epithelial cells. The inhibition of breast cancer cell growth was due to an induction of apoptosis, without any change in cell cycle progression. MCF-7 cells are known to express a wild-type p53 whereas MDA-MB-231 cells express a mutated p53. The fact that EGC induced apoptosis in both these cell lines suggests that the EGC-triggered apoptosis is independent of p53 status. Moreover, neutralizing antibodies against the death receptor Fas and inhibitors of caspases, such as caspase-8 and -10, efficiently inhibited the EGC-triggered apoptosis. In addition, immunoblotting revealed that EGC treatment was correlated with a decrease in Bcl-2 and an increase in Bax level. These results suggest that EGC-triggered apoptosis in breast cancer cells requires Fas signaling.


Subject(s)
Apoptosis/drug effects , Breast Neoplasms/metabolism , Catechin/analogs & derivatives , Catechin/pharmacology , Proto-Oncogene Proteins c-bcl-2 , Breast/cytology , Breast/metabolism , Breast Neoplasms/genetics , Caspases/metabolism , Cell Cycle/drug effects , Cyclin D1/analysis , Dose-Response Relationship, Drug , Fatty Acid Synthases/metabolism , Female , Humans , Proto-Oncogene Proteins/analysis , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/metabolism , bcl-2-Associated X Protein
13.
Breast Cancer Res Treat ; 71(3): 269-80, 2002 Feb.
Article in English | MEDLINE | ID: mdl-12002345

ABSTRACT

Cancer development depends not only on the nature of cancerous cells themselves, but also on the regulatory effects of various normal cells. The present study was performed to investigate the effect of normal breast epithelial cells (NBEC) on the growth of breast cancer cells under various conditions. We demonstrated that NBEC-conditioned medium (NBEC-CM) inhibited growth of breast cancer cell lines in monolayer culture and three-dimensional collagen gel culture, as well as in soft agar. In MCF-7 and T-47D cells which have a functional p53, NBEC-CM induced apoptosis without modifying cell cycle progression. In MDA-MB-231 and BT-20 cells that have a non-functional p53, NBEC-CM did not induce apoptosis, although a slight G1 blokage was observed in MDA-MB-231 cells. Transient transfections of MCF-7 and T-47D cells demonstrated that NBEC-triggered apoptosis was mediated by endogenous p53. Moreover, pifithrin-alpha which specifically inhibits the transcriptional activity of p53, completely abolished NBEC-induced apoptosis in both MCF-7 and T-47D cells, indicating that p53 mediated apoptosis via its transcriptional activity. Finally, orthovanadate, a protein tyrosine phosphatase inhibitor, completely inhibited NBEC-triggered apoptosis, indicating that NBEC-triggered apoptosis was regulated by tyrosine phosphatases.


Subject(s)
Apoptosis/drug effects , Breast Neoplasms/pathology , Breast/metabolism , Cell Cycle/drug effects , Epithelial Cells/metabolism , Tumor Suppressor Protein p53/genetics , Blotting, Western , Breast/cytology , Breast Neoplasms/metabolism , Cells, Cultured , Coculture Techniques , Culture Media, Conditioned/pharmacology , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Tumor Cells, Cultured
14.
Br J Pharmacol ; 135(1): 79-86, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11786482

ABSTRACT

1. This study was performed to determine the effect and action mechanisms of sodium butyrate (NaB) on the growth of breast cancer cells. 2. Butyrate inhibited the growth of all breast cancer cell lines analysed. It induced cell cycle arrest in G1 and apoptosis in MCF-7, MCF-7ras, T47-D, and BT-20 cells, as well as arrest in G2/M in MDA-MB-231 cells. 3. Transient transfection of MCF-7 and T47-D cells with wild-type and antisense p53 did not modify butyrate-induced apoptosis. Pifithrin-alpha, which inhibits the transcriptional activity of P53, did not modify cell growth or apoptosis of MCF-7 and T47-D cells treated with butyrate. These results indicate that P53 was not involved in butyrate-induced growth inhibition of breast cancer cells. 4. Treatment of MCF-7 cells with anti-Fas agonist antibody induced cell death, indicating that Fas was functional in these cells. Moreover, butyrate potentiated Fas-induced apoptosis, as massive apoptosis was observed rapidly when MCF-7 cells were treated with butyrate and anti-Fas agonist antibody. In addition, butyrate-induced apoptosis in MCF-7 cells was considerably reduced by anti-Fas antagonist antibody. Western blot analysis showed that butyrate increased Fas and Fas ligand levels (Fas L), indicating that butyrate-induced apoptosis may be mediated by Fas signalling. 5. These results demonstrate that butyrate inhibited the growth of breast cancer cells in a P53-independent manner. Moreover, it induced apoptosis via the Fas/Fas L system and potentiated Fas-triggered apoptosis in MCF-7 cells. These findings may open interesting perspectives in human breast cancer treatment strategy.


Subject(s)
Apoptosis/drug effects , Breast Neoplasms/metabolism , Butyrates/pharmacology , Tumor Suppressor Protein p53/physiology , fas Receptor/metabolism , Apoptosis/physiology , Benzothiazoles , Blotting, Western , Breast Neoplasms/physiopathology , Caspase Inhibitors , Cell Cycle/drug effects , Cell Division/drug effects , Fas Ligand Protein , Humans , Membrane Glycoproteins/drug effects , Membrane Glycoproteins/metabolism , Proto-Oncogene Proteins/drug effects , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism , Signal Transduction , Thiazoles/pharmacology , Toluene/analogs & derivatives , Toluene/pharmacology , Tumor Cells, Cultured/drug effects , bcl-2-Associated X Protein , fas Receptor/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...