Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters











Publication year range
1.
Life Sci ; 307: 120853, 2022 Oct 15.
Article in English | MEDLINE | ID: mdl-35926589

ABSTRACT

AIMS: Radioresistance and recurrences are crucial hindrances in cancer radiotherapy. Fractionated irradiation can elicit a mesenchymal phenotype in irradiated surviving cells and a deep connection exists between epithelial mesenchymal transition, radioresistance, and metastasis. The aim of this study was to analyze the effect of the secretoma of irradiated non-tumorigenic mammary epithelial cells on surviving irradiated breast tumor cells regarding the gain of mesenchymal traits and migratory ability. MAIN METHODS: MDA-MB-231 and MCF-7 breast cancer cells, irradiated or not, were incubated with conditioned media from MCF-10A non-tumorigenic epithelial breast cells, irradiated or not. After five days, we evaluated the expression and localization of epithelial and mesenchymal markers (by western blot and indirect immunofluorescence), cell migration (using transwells) and metalloproteinases activity (by zymography). We also assessed TGF-ß1 content in conditioned media by immunoblot, and the effect of A83-01 (a selective inhibitor of TGF-ß receptor I) and PP2 (a Src-family tyrosine kinase inhibitor) on nuclear Slug and cell migration. KEY FINDINGS: Conditioned media from MCF-10A cells caused phenotypic changes in breast tumor cells with attainment or enhancement of mesenchymal traits mediated at least in part by the activation of the TGF-ß type I receptor and a signaling pathway involving Src activation/phosphorylation. The effects were more pronounced mostly in irradiated tumor cells treated with conditioned media from irradiated MCF-10A. SIGNIFICANCE: Our results suggest that non-tumorigenic epithelial mammary cells included in the irradiation field could affect the response to irradiation of post-surgery residual cancer cells enhancing EMT progression and thus modifying radiotherapy efficacy.


Subject(s)
Neoplasms , Transforming Growth Factor beta1 , Cell Line, Tumor , Cell Movement , Culture Media, Conditioned/pharmacology , Epithelial Cells/metabolism , Epithelial-Mesenchymal Transition , Humans , MCF-7 Cells , Metalloproteases , Phenotype , Radiation, Ionizing , Receptor, Transforming Growth Factor-beta Type I , Transforming Growth Factor beta1/metabolism , src-Family Kinases
2.
Biochem Pharmacol ; 180: 114177, 2020 10.
Article in English | MEDLINE | ID: mdl-32721509

ABSTRACT

Epithelial-mesenchymal transition (EMT) contributes to cell invasion and metastasis during the progression of epithelial cancers. Though preclinical evidence suggests a role for histamine H4 receptor (H4R) in breast cancer growth, its function in the EMT is less known. In this study we proposed to investigate the effects of H4R ligands on EMT and mammosphere formation as a surrogate assay for cancer stem cells in breast cancer cells with different invasive phenotype. We also investigated the participation of Src and TGF-ß signaling in these events. Breast cancer cells were treated with the H4R agonists Clobenpropit, VUF8430 and JNJ28610244 and the H4R antagonist JNJ7777120. Immunodetection studies showed cytoplasmic E-cadherin, cytoplasmic and nuclear beta-catenin, nuclear Slug and an increase in vimentin and α-smooth muscle actin expression. There was also an enhancement in cell migration and invasion assessed by transwell units. All these effects were prevented by JNJ7777120. Moreover, H4R agonists induced an increase in phospho-Src levels detected by Western blot. Results revealed the involvement of phospho-Src in EMT events. Upon treatment with H4R agonists there was an increase in phospho-ERK1/2 and TGF-ß1 levels by Western blot, in Smad2/3 positive nuclei by indirect immunofluorescence, and in tumor spheres formation by the mammosphere assay. Notably, the selective TGF-ß1 kinase/activin receptor-like kinase inhibitor A83-01 blocked these effects. Moreover, cells derived from mammospheres exhibited higher Slug expression and enhanced migratory behavior. Collectively, findings support the interaction between H4R and TGF-ß receptor signaling in the enhancement of EMT features and mammosphere formation and point out intracellular TGF-ß1 as a potential mediator of these events.


Subject(s)
Breast Neoplasms/metabolism , Epithelial-Mesenchymal Transition/physiology , Oncogene Protein pp60(v-src)/metabolism , Receptors, Histamine H4/agonists , Receptors, Histamine H4/metabolism , Transforming Growth Factor beta1/metabolism , Epithelial-Mesenchymal Transition/drug effects , Female , Humans , Indoles/pharmacology , MCF-7 Cells , Piperazines/pharmacology
3.
Oncotarget ; 8(16): 26471-26491, 2017 Apr 18.
Article in English | MEDLINE | ID: mdl-28460440

ABSTRACT

The aims of the work were to improve our knowledge of the role of H4R in melanoma proliferation and assess in vivo the therapeutic efficacy of histamine, clozapine and JNJ28610244, an H4R agonist, in a preclinical metastatic model of melanoma. Additionally, we aimed to investigate the combinatorial effect of histamine and gamma radiation on the radiobiological response of melanoma cells.Results indicate that 1205Lu metastatic melanoma cells express H4R and that histamine inhibits proliferation, in part through the stimulation of the H4R, and induces cell senescence and melanogenesis. Daily treatment with H4R agonists (1 mg/kg, sc) exhibited a significant in vivo antitumor effect and importantly, compounds reduced metastatic potential, particularly in the group treated with JNJ28610244, the H4R agonist with higher specificity. H4R is expressed in benign and malignant lesions of melanocytic lineage, highlighting the potential clinical use of histamine and H4R agonists. In addition, histamine increased radiosensitivity of melanoma cells in vitro and in vivo. We conclude that stimulation of H4R by specific ligands may represent a novel therapeutic strategy in those tumors that express this receptor. Furthermore, through increasing radiation-induced response, histamine could improve cancer radiotherapy for the treatment of melanoma.


Subject(s)
Antineoplastic Agents/pharmacology , Histamine/pharmacology , Melanoma/metabolism , Melanoma/pathology , Animals , Antineoplastic Agents/therapeutic use , Biomarkers , Cell Line, Tumor , Cell Proliferation/drug effects , Cellular Senescence/drug effects , Combined Modality Therapy , Disease Models, Animal , Histamine/therapeutic use , Humans , Immunohistochemistry , Indoles/pharmacology , Melanoma/therapy , Mice , Mice, Nude , Neoplasm Metastasis , Neoplasm Staging , Oximes/pharmacology , Radiation, Ionizing , Receptors, Histamine H4/metabolism , Xenograft Model Antitumor Assays
4.
Pharmacol Res ; 111: 731-739, 2016 09.
Article in English | MEDLINE | ID: mdl-27473821

ABSTRACT

Radiotherapy is a prime option for treatment of solid tumors including breast cancer though side effects are usually present. Experimental evidence shows an increase in invasiveness of several neoplastic cell types through conventional tumor irradiation. The induction of epithelial to mesenchymal transition is proposed as an underlying cause of metastasis triggered by gamma irradiation. Experiments were conducted to investigate the role of histamine on the ionizing radiation-induced epithelial to mesenchymal transition events in breast cancer cells with different invasive phenotype. We also evaluated the potential involvement of Src phosphorylation in the migratory capability of irradiated cells upon histamine treatment. MCF-7 and MDA-MB-231 mammary tumor cells were exposed to a single dose of 2Gy of gamma radiation and five days after irradiation mesenchymal-like phenotypic changes were observed by optical microscope. The expression and subcellular localization of E-cadherin, ß-catenin, vimentin and Slug were determined by immunoblot and indirect immunofluorescence. There was a decrease in the epithelial marker E-cadherin expression and an increase in the mesenchymal marker vimentin after irradiation. E-cadherin and ß-catenin were mainly localized in cytoplasm. Slug positive nuclei, matrix metalloproteinase-2 activity and cell migration and invasion were significantly increased. In addition, a significant enhancement in Src phosphorylation/activation could be determined by immunoblot in irradiated cells. MCF-7 and MDA-MB-231 cells also received 1 or 20µM histamine during 24h previous to be irradiated. Notably, pre-treatment of breast cancer cells with 20µM histamine prevented the mesenchymal changes induced by ionizing radiation and also reduced the migratory behavior of irradiated cells decreasing phospho-Src levels. Collectively, our results suggest that histamine may block events related to epithelial to mesenchymal transition in irradiated mammary cancer cells and open a perspective for the potential use of histamine to improve radiotherapy efficacy.


Subject(s)
Breast Neoplasms/radiotherapy , Cell Movement/drug effects , Epithelial-Mesenchymal Transition/drug effects , Histamine/pharmacology , Radiation-Protective Agents/pharmacology , Antigens, CD , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cadherins/metabolism , Cell Movement/radiation effects , Dose-Response Relationship, Drug , Epithelial-Mesenchymal Transition/radiation effects , Female , Humans , MCF-7 Cells , Neoplasm Invasiveness , Phenotype , Phosphorylation , Radiotherapy/adverse effects , Signal Transduction/drug effects , Signal Transduction/radiation effects , Snail Family Transcription Factors/metabolism , Vimentin/metabolism , beta Catenin/metabolism , src-Family Kinases/metabolism
5.
Int J Biochem Cell Biol ; 51: 29-38, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24685678

ABSTRACT

Epithelial to mesenchymal transition (EMT) of cancer cells is an essential process in cancer progression. Cancer cells that undergone EMT loose cell-cell contacts, acquire mesenchymal properties and develop migratory and invasive abilities. In previous studies we have demonstrated that histamine may modify the invasive phenotype of pancreatic and mammary tumor cells. In this work we proposed to investigate whether histamine may also influence the interaction between tumor cells and normal fibroblasts. The potential activation of normal CCD-1059Sk fibroblasts by histamine and EMT phenotypic changes induced in MCF-7 and MDA-MB-231 breast tumor cells by the conditioned media (CM) derived from fibroblasts were evaluated. Initially, we determined the presence of H1, H2 and H4 histamine receptors and matrix metalloproteinase 2 (MMP2) mRNA in CCD-1059Sk fibroblasts. MMP2 gelatinolytic activity, cell migration and alpha-smooth muscle actin expression were increased in fibroblasts by low doses (<1µM) and decreased by high doses (20µM) of histamine. MCF-7 cells cultured with CM from fibroblasts exhibited spindle-shaped morphology, cell spreading and cytoplasmic expression of ß-catenin but there was no change in MMP2 activity and cell migration. MDA-MB-231 cells cultured with CM from fibroblasts showed a more elongated phenotype, cell spreading, cytoplasmic ß-catenin, increased MMP2 activity and endogenous TGF-ß1 expression, and enhanced cell migration and invasion. Notably, all these features were reversed when mammary tumor cells were cultured with CM from fibroblasts treated with 20µM histamine. In conclusion, high doses of histamine may prevent the activation of fibroblasts and also avert the EMT related changes induced in epithelial tumor cells by fibroblasts CM.


Subject(s)
Breast Neoplasms/pathology , Fibroblasts/metabolism , Histamine/pharmacology , Breast Neoplasms/therapy , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/physiology , Disease Progression , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/physiology , Female , Fibroblasts/cytology , Humans , MCF-7 Cells
6.
Biol Trace Elem Res ; 157(2): 138-46, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24363240

ABSTRACT

Scleroderma, sclerosis of the skin, is a severe autoimmune disease refractant to all kind of treatments. To study the in vivo effects of a combination of three oligoelements selenium (Se), zinc (Zn), and manganese (Mn) plus Lachesis muta venom (O-LM) on the bleomycin (BLM)-induced scleroderma mouse experimental model. C3H mice were randomly divided into four groups: control (phosphate-buffered saline (PBS)), O-LM, BLM, and BLM + O-LM. All administrations were performed subcutaneously into the back of mice. BLM was injected 5 days per week for three consecutive weeks and O-LM was administered simultaneously with BLM from the beginning of the experiments and lasted for 3 weeks after the final BLM or PBS injection (for O-LM and BLM + O-LM groups), when animals were sacrificed and histopathological, immunohistochemical, thiobarbituric acid reactive species (TBARS) evaluation, and autoantibodies detection were determined. O-LM significantly reduced BLM-induced enhanced dermal thickness (605 ± 47 vs. 956 ± 59 µm, P < 0.01), collagen deposition, and mast cells infiltration (43.1 ± 1.0 vs. 102 ± 14.1 mast cells, P < 0.05). O-LM administration significantly blocked BLM-induced oxidative damage and the enhanced immunoreactive fibroblasts for α-smooth muscle actin while reduced BLM-induced autoantibodies that strongly react mainly with skin and spleen. O-LM significantly reduced BLM-induced scleroderma through the modulation of antioxidant and immunological pathways.


Subject(s)
Crotalid Venoms/therapeutic use , Manganese/therapeutic use , Scleroderma, Systemic/drug therapy , Selenium/therapeutic use , Skin/drug effects , Zinc/therapeutic use , Animals , Antioxidants/metabolism , Autoantibodies/blood , Bleomycin/pharmacology , Cell Count , Cell Survival/drug effects , Crotalid Venoms/administration & dosage , Crotalid Venoms/toxicity , Disease Models, Animal , Drug Therapy, Combination , Manganese/administration & dosage , Manganese/toxicity , Mast Cells/drug effects , Mast Cells/immunology , Mast Cells/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Organ Specificity , Oxidative Stress/drug effects , Scleroderma, Systemic/immunology , Scleroderma, Systemic/pathology , Selenium/administration & dosage , Selenium/toxicity , Skin/immunology , Skin/pathology , Zinc/administration & dosage , Zinc/toxicity
7.
J Dermatol Sci ; 72(3): 252-62, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23999004

ABSTRACT

BACKGROUND: Functional presence of histamine H4 receptor (H4R) was demonstrated in human melanoma cell lines and biopsies. OBJECTIVE: The purposes of this work were to investigate signal transduction pathways and biological responses triggered by the activation of H4R in human primary (WM35) and metastatic (M1/15) melanoma cell lines and to evaluate the in vivo antitumor activity of histamine (HA) and clozapine (CLZ) on human M1/15 melanoma xenografts. METHODS: Clonogenic assay, incorporation of BrdU, cell cycle distribution, phosphorylation levels of ERK1/2 and cAMP production were evaluated in vitro. An experimental human melanoma model was developed into athymic nude mice. Tumor growth, survival and histochemical studies were performed in order to investigate the expression levels of H4R, HA, PCNA, mitotic index (MI), and angiogenesis. RESULTS: The results indicate that H4R agonists inhibited forskolin-induced cAMP levels only in M1/15 cells while increased phosphorylation levels of ERK1/2 and decreased proliferation in both cell types. In vivo studies show that HA and CLZ (1mgkg(-1), sc) significantly increased median survival and decreased tumor volume. These effects were associated to a reduction in MI, in the expression of proliferation marker and in intratumoral neovascularization. CONCLUSIONS: We conclude that HA and CLZ exhibit an antitumoral effect in vitro and in vivo on human melanoma, suggesting the therapeutic potential of these compounds for the treatment of malignant melanoma.


Subject(s)
Clozapine/therapeutic use , Histamine/therapeutic use , Melanoma, Experimental/drug therapy , Receptors, G-Protein-Coupled/agonists , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Clozapine/pharmacology , Cyclic AMP/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Histamine/pharmacology , Humans , Male , Melanoma, Experimental/metabolism , Mice , Mice, Nude , Neovascularization, Pathologic/drug therapy , Phosphorylation/drug effects , Receptors, Histamine , Receptors, Histamine H4 , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
8.
BMC Pharmacol Toxicol ; 14: 6, 2013 Jan 11.
Article in English | MEDLINE | ID: mdl-23311706

ABSTRACT

BACKGROUND: Glibenclamide (Gli) binds to the sulphonylurea receptor (SUR) that is a regulatory subunit of ATP-sensitive potassium channels (KATP channels). Binding of Gli to SUR produces the closure of KATP channels and the inhibition of their activity. This drug is widely used for treatment of type 2-diabetes and it has been signaled as antiproliferative in several tumor cell lines. In previous experiments we demonstrated the antitumoral effect of Gli in mammary tumors induced in rats. The aim of the present work was to investigate the effect of Gli on MDA-MB-231 breast cancer cell proliferation and to examine the possible pathways involved in this action. RESULTS: The mRNA expression of the different subunits that compose the KATP channels was evaluated in MDA-MB-231 cells by reverse transcriptase-polymerase chain reaction. Results showed the expression of mRNA for both pore-forming isoforms Kir6.1 and Kir6.2 and for the regulatory isoform SUR2B in this cell line. Gli inhibited cell proliferation assessed by a clonogenic method in a dose dependent manner, with an increment in the population doubling time. The KATP channel opener minoxidil increased clonogenic proliferation, effect that was counteracted by Gli. When cell cycle analysis was performed by flow cytometry, Gli induced a significant cell-cycle arrest in G0/G1 phase, together with an up-regulation of p27 levels and a diminution in cyclin E expression, both evaluated by immunoblot. However, neither differentiation evaluated by neutral lipid accumulation nor apoptosis assessed by different methodologies were detected. The cytostatic, non toxic effect on cell proliferation was confirmed by removal of the drug.Combination treatment of Gli with tamoxifen or doxorubicin showed an increment in the antiproliferative effect only for doxorubicin. CONCLUSIONS: Our data clearly demonstrated a cytostatic effect of Gli in MDA-MB-231 cells that may be mediated through KATP channels, associated to the inhibition of the G1-S phase progression. In addition, an interesting observation about the effect of the combination of Gli with doxorubicin leads to future research for a potential novel role for Gli as an adjuvant in breast cancer treatment.


Subject(s)
Antineoplastic Agents/pharmacology , Glyburide/pharmacology , Apoptosis/drug effects , Breast Neoplasms/drug therapy , Cell Line, Tumor , Cell Proliferation/drug effects , Female , G1 Phase/drug effects , Humans , Hypoglycemic Agents/pharmacology , KATP Channels/genetics , Membrane Potential, Mitochondrial/drug effects , RNA, Messenger/metabolism , Resting Phase, Cell Cycle/drug effects
9.
Biochem Cell Biol ; 90(6): 779-90, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23194191

ABSTRACT

Radiotherapy may be used to treat pancreatic cancer and relieve pain. We have previously reported that histamine modulates pancreatic adenocarcinoma PANC-1 cell proliferation. This work was aimed to evaluate whether histamine improves radiosensitivity of PANC-1 cells in relation to phosphorylation/inhibition of glycogen synthase kinase-3ß (GSK-3ß). Immediately after γ irradiation, intracellular hydrogen peroxide was markedly decreased together with a rapid increase in catalase activity. Although histamine diminished catalase activity in nonirradiated cells, it only partially hindered the increase observed in irradiated cells and could not modify radiosensitivity. In control cells, a high expression of total and a very low expression of phosphorylated/inactive GSK-3ß were found. An increment in reactive oxygen species levels produced an augmentation in GSK-3ß phosphorylation and suppressed cell proliferation. In both control and histamine-treated irradiated cells, the rise in catalase activity lowered reactive oxygen species levels and only a small increase in phosphorylated GSK-3ß was detected. Alternatively, 3-aminotriazole, an irreversible inhibitor of catalase, reduced the survival fraction in irradiated control cells along with an increment in phosphorylated GSK-3ß. These results suggest that upon irradiation, early catalase activation may be responsible for keeping GSK-3ß active conceding cells a survival advantage toward cytotoxic effects of ionizing radiation.


Subject(s)
Cell Proliferation/radiation effects , Glycogen Synthase Kinase 3/metabolism , Adenocarcinoma , Apoptosis , Cell Line, Tumor , Gamma Rays , Glycogen Synthase Kinase 3 beta , Humans , Pancreatic Neoplasms , Phosphorylation
10.
Melanoma Res ; 21(5): 395-404, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21691231

ABSTRACT

We have previously reported that histamine at micromolar concentrations reduces the proliferation of melanoma cell lines. It is also known that melanoma cells express histamine H1, H2, and H3 receptors. The aim of this study was to investigate the presence of histamine H4 receptor (H4R) in human melanoma cells and its associated biological processes. To better understand the importance of histamine in tumor development, we explored the expression of H4R in human melanoma tissue biopsies. The expression of H4R in WM35 and M1/15 cells was analyzed by reverse-transcription-PCR, western blot, and immunocytochemistry. To characterize the biological responses we evaluated cell proliferation by clonogenic assay and 5-bromo-2'-deoxyuridine incorporation. In addition, cell senescence and differentiation were determined by ß-galactosidase enzyme assay and dopa oxidase activity, respectively. The expression levels of H4R were determined by immunohistochemistry in 19 samples of human malignant lesions. Results indicate that melanoma cells express H4R at the messenger RNA and protein levels. By using histamine agonists, antagonists, and H4R small-interfering RNA we showed that the inhibitory effect of histamine on proliferation was in part mediated through the stimulation of the H4R. The decrease in proliferation was associated with an induction of cell senescence and an increase in melanogenesis, which is a differentiation marker of these cells. Furthermore, H4R was expressed in 42% of human melanoma biopsies. To our knowledge, this is the first report that describes the presence of the H4R in melanoma cells and tissue, suggesting a potential therapeutic application of H4R ligands.


Subject(s)
Histamine/pharmacology , Melanoma/metabolism , Receptors, G-Protein-Coupled/biosynthesis , Receptors, Histamine/biosynthesis , Skin Neoplasms/metabolism , Cell Differentiation/physiology , Cell Growth Processes/physiology , Cell Line, Tumor , Guanidines/pharmacology , Histamine/metabolism , Humans , Imidazoles/pharmacology , Immunohistochemistry , Indoles/pharmacology , Melanoma/genetics , Melanoma/pathology , Piperazines/pharmacology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/genetics , Receptors, Histamine/genetics , Receptors, Histamine H4 , Thiourea/analogs & derivatives , Thiourea/pharmacology
11.
Free Radic Biol Med ; 46(11): 1510-5, 2009 Jun 01.
Article in English | MEDLINE | ID: mdl-19285550

ABSTRACT

Histamine is a recognized growth factor in melanoma, and exogenous histamine produces a dual effect on proliferation. We have previously reported that histamine at micromolar concentrations reduces the proliferation of melanoma cell lines. To investigate the mechanism by which histamine inhibits proliferation of WM35 human melanoma cells, we have studied the involvement of histamine in reactive oxygen species production and antioxidant enzyme regulation in these cells. Results indicate that histamine treatment (10 microM) significantly increased hydrogen peroxide levels, whereas it slightly decreased superoxide levels associated with an enhancement of superoxide dismutase and a reduction in catalase activity. Additionally, catalase treatment reversed the inhibitory effect of histamine on proliferation, and various treatments that reduce hydrogen peroxide formation increased proliferation of these cells. Furthermore, we demonstrate that the inhibition of proliferation produced by histamine was mediated at least in part by an induction of cell senescence. We conclude that hydrogen peroxide is involved in histamine-mediated modulation of proliferation in malignant melanoma cells.


Subject(s)
Cell Proliferation , Enzyme Activation/immunology , Histamine/metabolism , Hydrogen Peroxide/metabolism , Melanoma/pathology , Catalase/metabolism , Cellular Senescence/immunology , Histamine/immunology , Humans , Melanoma/enzymology , Reactive Oxygen Species/metabolism , Superoxide Dismutase/metabolism
12.
World J Gastroenterol ; 15(9): 1065-71, 2009 Mar 07.
Article in English | MEDLINE | ID: mdl-19266598

ABSTRACT

AIM: To study the action of aminoguanidine on pancreatic cancer xenografts in relation to cell proliferation, apoptosis, redox status and vascularization. METHODS: Xenografts of PANC-1 cells were developed in nude mice. The animals were separated into two groups: control and aminoguanidine treated. Tumor growth, survival and appearance of metastases were determined in vivo in both groups. Tumors were excised and ex vivo histochemical studies were performed. Cell growth was assessed by Ki-67 expression. Apoptosis was studied by intratumoral expression of B cell lymphoma-2 protein (Bcl-2) family proteins and Terminal deoxynucleotidyl transferase biotin-dUTP Nick End Labeling (Tunel). Redox status was evaluated by the expression of endothelial nitric oxide synthase (eNOS), catalase, copper-zinc superoxide dismutase (CuZnSOD), manganese superoxide dismutase (MnSOD) and glutathione peroxidase (GPx). Finally, vascularization was determined by Massons trichromic staining, and by VEGF and CD34 expression. RESULTS: Tumor volumes after 32 d of treatment by aminoguanidine (AG) were significantly lower than in control mice (P < 0.01). Median survival of AG mice was significantly greater than control animals (P < 0.01). The appearance of both homolateral and contralateral palpable metastases was significantly delayed in AG group. Apoptotic cells, intratumoral vascularization (trichromic stain) and the expression of Ki-67, Bax, eNOS, CD34, VEGF, catalase, CuZnSOD and MnSOD were diminished in AG treated mice (P < 0.01), while the expression of Bcl-2 and GPx did not change. CONCLUSION: The antitumoral action of aminoguanidine is associated with decreased cell proliferation, reduced angiogenesis, and reduced expression of antioxidant enzymes.


Subject(s)
Cell Division/drug effects , Guanidines/therapeutic use , Neoplasm Metastasis/prevention & control , Neoplasm Transplantation/methods , Pancreatic Neoplasms/pathology , Transplantation, Heterologous , Animals , Antigens, CD34/genetics , Enzyme Inhibitors/therapeutic use , Glutathione Peroxidase/genetics , Humans , Mice , Mice, Nude , Nitric Oxide Synthase Type III/genetics , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/enzymology , Superoxide Dismutase/genetics , Vascular Endothelial Growth Factor A/genetics
13.
Cancer Biol Ther ; 7(1): 28-35, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17932461

ABSTRACT

There is increasing evidence that describes a histamine role in normal and cancer cell proliferation. To better understand the importance of histamine in breast cancer development, the expression of histamine H3 (H3R) and H4 (H4R) receptors and their association with proliferating cell nuclear antigen (PCNA), histidine decarboxylase (HDC) and histamine content were explored in mammary biopsies. Additionally, we investigated whether H3R and H4R were implicated in the biological responses triggered by histamine in MDA-MB-231 breast cancer cells. The expression levels of H3R, H4R, PCNA, HDC and histamine content were determined by immunohistochemistry in 40 benign and malignant lesions. MDA-MB-231 cells proliferation (clonogenic assay and BrdU incorporation) and cell cycle distribution (flow cytometry) were evaluated upon treatment with histamine, H3R and H4R agonists and antagonists. Apoptosis was determined by Annexin staining and TUNEL assay. Cell migration was assessed by transwell system. Results indicate that H3R was detected in 67% (10/15) of benign lesions and in almost all carcinomas (24/25), being the level of its expression significantly higher in carcinomas (p = 0.0016). The non-tumoral breast tissue surrounding carcinomas revealed a lower H3R expression compared to the tumor cells. Only 13% (2/15) of the benign lesions expressed H4R compared to 44% (11/25) of the carcinomas. Interestingly, H3R expression was correlated in carcinomas with the expression of HDC and PCNA (p < 0.0001), and also histamine content (p = 0.0229). Accordingly, histamine increased MDA-MB-231 cells proliferation and also migration via H3R. In contrast, activation of H4R inhibited proliferation and this effect was associated with an arrest in the G(0)/G(1) phase of the cell cycle and an induction of apoptosis. Present findings demonstrate the presence of H3R and H4R in human mammary tissue and suggest that H3R may be involved in the regulation of breast cancer growth and progression representing a novel molecular target for new therapeutic approach.


Subject(s)
Breast Neoplasms/etiology , Histamine/physiology , Receptors, G-Protein-Coupled/physiology , Receptors, Histamine H3/physiology , Receptors, Histamine/physiology , Adult , Aged , Breast/chemistry , Breast Neoplasms/drug therapy , Cell Movement/drug effects , Cell Proliferation/drug effects , Female , Histamine/analysis , Histidine Decarboxylase/analysis , Humans , Imidazoles/pharmacology , Middle Aged , Proliferating Cell Nuclear Antigen/analysis , Receptors, G-Protein-Coupled/analysis , Receptors, G-Protein-Coupled/drug effects , Receptors, Histamine/analysis , Receptors, Histamine/drug effects , Receptors, Histamine H3/analysis , Receptors, Histamine H3/drug effects , Receptors, Histamine H4 , Thiourea/analogs & derivatives , Thiourea/pharmacology
14.
Int J Radiat Biol ; 83(10): 653-63, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17729160

ABSTRACT

PURPOSE: To examine the protective effects of histamine on intestinal damage produced by gamma-radiation. MATERIALS AND METHODS: 56 mice were divided into 4 groups. Histamine and Histamine-10 Gy groups received a daily subcutaneous histamine injection (0.1 mg/kg) starting 20 hours before irradiation and continued until the end of the experimental period; the untreated group received saline. Histamine-10 Gy and untreated-10 Gy groups were irradiated with a single dose on whole-body using Cesium-137 source (7 Gy/min) and were sacrificed 3 days after irradiation. Small intestine was removed, fixed and stained with hematoxylin and eosin. The number of intestinal crypts per circumference, and other histological characteristics of intestinal cells were evaluated. We further determined by immunohistochemistry the expression of proliferating cell nuclear antigen (PCNA), Bax, Bcl-2 (pro- and anti-apoptotic protein, respectively), antioxidant enzymes (Superoxide dismutase (SOD), Catalase and Glutathione peroxidase), histamine content and apoptosis by terminal deoxynucleotidyl transferase mediated deoxyuridine triphosphate biotin nick end labeling (TUNEL) assay. Cells in the S phase of the cell cycle were identified by immunohistochemical detection of 5-bromo-2'-deoxyuridine (BrdU) incorporation. RESULTS: Histamine treatment reduced mucosal atrophy, edema and preserved villi, crypts and nuclear and cytoplasmic characteristics of small intestine after radiation exposure. Additionally, histamine treatment increased PCNA expression and the BrdU-positive cell number, histamine content, decreased the number of apoptotic cells and significantly increased Catalase and copper-zinc-containing SOD of irradiated mice. CONCLUSIONS: Histamine prevents radiation-induced toxicity by increasing proliferation of damaged intestinal mucosa and suppressing apoptosis that was associated with an increase in SOD and Catalase levels. This effect might be of clinical value in patients undergoing radiotherapy.


Subject(s)
Apoptosis/drug effects , Cell Proliferation/drug effects , Cesium Isotopes/metabolism , Histamine/administration & dosage , Intestine, Small/drug effects , Radiation Injuries, Experimental/prevention & control , Animals , Cell Nucleus/pathology , Cytoplasm/pathology , Edema/pathology , Histamine/pharmacology , Immunohistochemistry , Injections, Subcutaneous , Intestinal Diseases/drug therapy , Intestinal Diseases/pathology , Intestinal Diseases/radiotherapy , Intestine, Small/pathology , Intestine, Small/radiation effects , Mice , Mice, Nude , Peroxidases/metabolism , Proliferating Cell Nuclear Antigen/metabolism , Radiation Injuries, Experimental/drug therapy , Radiation Injuries, Experimental/veterinary , Time Factors , Whole-Body Irradiation
15.
Cancer Biol Ther ; 5(11): 1462-71, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17012845

ABSTRACT

Histamine is a biogenic amine responsible for multiple biological actions including regulation of physiological functions of mammary gland. It has been postulated that histamine plays a critical role in proliferation of normal and cancer cells. To investigate the biological responses that histamine exerts in malignant cells derived from human mammary gland, we evaluated in MDA-MB-231 line the expression of histamine receptors, histamine intracellular content, the capacity of histamine to influence proliferation, cell cycle progression, differentiation and apoptosis. We also studied histamine involvement in cellular response to ionizing radiation. HBL-100 cells were used as control of non-tumorigenic breast cells. Proliferation and surviving fraction were assessed by clonogenic assay. Cell cycle progression and lipid accumulation were determined by flow cytometry while apoptosis was studied by Annexin V and DNA fragmentation assays. Both cell lines expressed the four histamine receptors subtypes as evaluated by western blot and RT-PCR analyses, and present endogenous histamine. Histamine regulated proliferation of cancer cells in a dose-dependent way and 10 microM histamine reduced significantly proliferation to 23% inducing cell cycle arrest in G(2)/M phase, differentiation by 26% and a significant increase in the number of apoptotic cells (p < 0.01). These responses were not observed in HBL-100 cells. Furthermore, 10 microM histamine exclusively enhanced the radiosensitivity of MDA-MB-231 cells. These results represent the first report about the expression of H3 and H4 receptors in human breast cells. In addition, we conclude that histamine exerts different effects on biological responses of normal and cancer breast cells representing a promising target for the development of more specific and less toxic cancer therapies.


Subject(s)
Breast Neoplasms/pathology , Breast Neoplasms/physiopathology , Histamine/pharmacology , Signal Transduction/physiology , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Division/drug effects , Cell Line, Tumor , DNA Primers , Female , Humans , RNA, Neoplasm/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects
16.
Anticancer Res ; 26(3A): 2113-22, 2006.
Article in English | MEDLINE | ID: mdl-16827153

ABSTRACT

The objective of this study was to evaluate the in vivo antitumor action of rosiglitazone (Rosi) alone or in combination with tamoxifen (Tam) on experimental mammary tumors induced by N-nitroso-N-methylurea (NMU) in Sprague-Dawley rats. Animals bearing mammary tumors were treated with 0.06 mg/kg/day or 0.12 mg/kg/day of Rosi orally, 1 mg/kg/day of Tam s.c., or with the combined treatment (Rosi+Tam). After 25 days of treatment, the following responses were observed: 45% of tumors were responsive to 0.06 mg/kg/day of Rosi treatment, while 55% of tumors under Tam treatment responded. The results of the combined Rosi+Tam treatment indicated that 75% of tumors were responsive. Similar results were obtained with 0.12 mg/kg/day of Rosi. Apoptosis, necrosis and glandular hypersecretion were observed in Rosi-treated tumors. In all cases, the combined Rosi+Tam treatment potentiated the antitumor effect of Tam alone. No side-effects were observed after treatment at any assayed dose.


Subject(s)
Mammary Neoplasms, Experimental/drug therapy , Thiazolidinediones/pharmacology , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Body Weight/drug effects , Cell Growth Processes/drug effects , Drinking/drug effects , Eating/drug effects , Female , Glucose Tolerance Test , Insulin/blood , Insulin-Like Growth Factor I/metabolism , Mammary Neoplasms, Experimental/chemically induced , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Methylnitrosourea , Rats , Rats, Sprague-Dawley , Rosiglitazone , Tamoxifen/administration & dosage , Thiazolidinediones/administration & dosage
17.
Anticancer Res ; 26(6B): 4443-50, 2006.
Article in English | MEDLINE | ID: mdl-17201167

ABSTRACT

In this study, the mechanisms involved in the inhibitory effect of histamine (HA) on PANC-1 cell proliferation were investigated. The action of HA on cell growth was evaluated by determining the cell doubling time from experimental growth curves and analysing the cell cycle using a flow cytometer. The expression of proteins related to cell death and proliferation (PCNA, p53, c-Fos and Bcl-2 family proteins) was studied using Western blot, immunocytochemistry and flow cytometric analysis. The results indicated that HA produced an accumulation of PANC-1 cells in GO/Gl-phase and increased the doubling time via H2HA (H2R) stimulation. Expression of p53, c-Fos and Bcl-2 were not modulated by HA. However, HA decreased PCNA and Bax expression, while it increased the Bcl-x level. In summary, the antiproliferative effect exerted by HA was associated with a G0/G1-phase arrest and a modulation of the Bcl-2 family proteins.


Subject(s)
Cell Proliferation/drug effects , Histamine/pharmacology , Proto-Oncogene Proteins c-bcl-2/physiology , Receptors, Histamine H2/physiology , Cell Death/drug effects , Cell Line, Tumor , Flow Cytometry , Humans , Immunohistochemistry
18.
Anticancer Res ; 25(6B): 4305-12, 2005.
Article in English | MEDLINE | ID: mdl-16309233

ABSTRACT

The aim of this study was to investigate the expression and localization of the insulin growth factor type 1 receptor (IGF-IR) in malignant and benign mammary tumors induced in rats by N-nitroso-N-methylurea (NMU) and its correlation with histopathology and hormone dependence. Also, protein tyrosine kinase activities (PTKs) were analyzed in order to study the activation of the intracellular cascade. The results showed that IGF-IR is present in NMU tumors (analyzed by binding assay and Western blot), that a variable content is expressed in tumors that continued growing post-ovariectomy (OVX) of rats, and that it is undetectable in tumors that regressed post-OVX. IGF-IR was principally localized (by immunohistochemistry) in the epithelial cells of malignant tumors and in the fibrous cells of benign ones. Also, a significantly lower expression of both cytosolic and microsomal PTKs were found in benign tumors. Our results suggest a different expression and role of IGF-IR in benign and malignant tumors.


Subject(s)
Mammary Neoplasms, Experimental/metabolism , Receptor, IGF Type 1/metabolism , Animals , Blotting, Western , Disease Progression , Female , Immunohistochemistry , Mammary Neoplasms, Experimental/enzymology , Mammary Neoplasms, Experimental/pathology , Neoplasms, Hormone-Dependent/enzymology , Neoplasms, Hormone-Dependent/metabolism , Neoplasms, Hormone-Dependent/pathology , Protein-Tyrosine Kinases/metabolism , Radioligand Assay , Rats , Rats, Sprague-Dawley , Receptor, IGF Type 1/biosynthesis
19.
Biocell ; 29(2): 149-61, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16187493

ABSTRACT

Apoptosis or programmed cell death (PCD) is a physiological process characteristic of pluricellular organisms leading to self-destruction of the cell. It is therefore involved in development, homeostasis and host defense. However, a significant difference has been shown between mammalian cell apoptosis and non-mammalian cell apoptosis: mitochondria are implicated only in the former. Execution of PCD includes the release of several proapoptotic proteins from the intermembrane space of mitochondria. They could exert their actions through a caspase dependent as well as a caspase independent way. On the other hand, regulation of PCD is mainly given by the Bcl-2 family members, which are in turn essentially regulated by activation of death receptors and/or DNA damage. Nowadays, execution of apoptosis is better known than its regulation. Nevertheless, we are still far of a complete understanding of the apoptotic process.


Subject(s)
Apoptosis , Mitochondria/pathology , Animals , Caspases/metabolism , Cytochromes c/metabolism , DNA Damage , Endodeoxyribonucleases/metabolism , Enzyme Activation , Humans , Intracellular Membranes/metabolism , Mitochondria/metabolism , Models, Biological , Necrosis , Phosphorylation , Protein Structure, Tertiary , Proto-Oncogene Proteins c-bcl-2/metabolism
20.
Biocell ; Biocell;29(2): 149-161, ago. 2005. ilus
Article in English | LILACS | ID: lil-429669

ABSTRACT

Apoptosis or programmed cell death (PCD) is a physiological process characteristic of pluricellular organisms leading to self-destruction of the cell. It is therefore involved in development, homeostasis and host defense. However, a significant difference has been shown between mammalian cell apoptosis and non-mammalian cell apoptosis: mitochondria are implicated only in the former. Execution of PCD includes the release of several proapoptotic proteins from the intermembrane space of mitochondria. They could exert their actions through a caspase dependent as well as a caspase independent way. On the other hand, regulation of PCD is mainly given by the Bcl-2 family members, which are in turn essentially regulated by activation of death receptors and/or DNA damage. Nowadays, execution of apoptosis is better known than its regulation. Nevertheless, we are still far of a complete understanding of the apoptotic process


Subject(s)
Animals , Apoptosis/physiology , Cytochromes c , Ear Canal/cytology , Ear Canal/metabolism , Homeostasis , Mitochondria/physiology , Mitochondria/metabolism , Mammals/anatomy & histology , Nematoda/anatomy & histology , Nematoda/cytology
SELECTION OF CITATIONS
SEARCH DETAIL