Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Food Chem Toxicol ; 156: 112515, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34400204

ABSTRACT

Chlorpyrifos (CPF), the most used insecticide in Argentina, can act as an endocrine disruptor at low doses. We previously demonstrated that chronic exposure to CPF induces hormonal imbalance in vivo. The aim of this work was to study the effects of low concentrations of CPF (0.01 and 1 mg/kg/day) on the reproductive system of virgin adult rats. In the ovary, we studied the effects of CPF on steroidogenesis by determining steroid hormone content by RIA and CYP11 and CYP19 enzyme expression by qRT-PCR. The estrous cycle was evaluated by microscopic observation of vaginal smear, as well as by changes in uterine histology. In endometrium, we determined the fractal dimension and expression of PCNA, ERα and PR by IHC. Our results showed that chronic exposure to CPF affects ovarian steroid synthesis, causing alterations in the normal cyclicity of animals. In addition, CPF induced proliferative changes in the uterus, suggesting that it could affect reproduction or act as a risk factor in the development of uterine proliferative pathologies.


Subject(s)
Chlorpyrifos/administration & dosage , Chlorpyrifos/toxicity , Estrous Cycle/drug effects , Ovary/drug effects , Uterus/drug effects , Animals , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Insecticides/administration & dosage , Insecticides/toxicity , Random Allocation , Rats , Rats, Sprague-Dawley , Vagina/drug effects
2.
J Cell Physiol ; 235(11): 8757-8767, 2020 11.
Article in English | MEDLINE | ID: mdl-32324259

ABSTRACT

Metabolic reprogramming of cancer cells results in a high production of acidic substances that must be extruded to maintain tumor-cell viability. The voltage-gated proton channel (Hv1) mediates highly selective effluxes of hydronium-ion (H+ ) that prevent deleterious cytoplasmic acidification. In the work described here, we demonstrated for the first time that the amino-terminal-truncated isoform of Hv1 is more highly expressed in tumorigenic breast-cancer-cell lines than in nontumorigenic breast cells. With respect to Hv1 function, we observed that pharmacologic inhibition of that channel, mediated by the specific blocker 5-chloro-2-guanidinobenzimidazole, produced a drop in intracellular pH and a decrease in cell viability, both in monolayer and in three-dimensional cultures, and adversely affected the cell-cycle in tumorigenic breast cells without altering the cycling of nontumorigenic cells. In conclusion, our results demonstrated that the Hv1 channel could be a potential tool both as a biomarker and as a therapeutic target in breast-cancer disease.


Subject(s)
Breast Neoplasms/metabolism , Cell Survival/physiology , Ion Channels/metabolism , Humans , Hydrogen-Ion Concentration , Protein Isoforms/metabolism
3.
J Steroid Biochem Mol Biol ; 199: 105594, 2020 05.
Article in English | MEDLINE | ID: mdl-31968225

ABSTRACT

Breast cancer is currently the leading cause of cancer death among women worldwide. AP-1 (c-Fos/c-Jun) is associated with proliferation and survival, while cytoplasmic c-Fos activates phospholipid synthesis in cells induced to differentiate or grow. Estrogen receptor α 46 (ERα46) is a splice variant of full-length ERα66 and it is known that it has an inhibitory role in cancer cell growth. We investigated c-Fos localization, its relationship to AP-1, the non genomic pathway of phospho-Tyr537-ERα66, as well as ERα46 and ERα66 isoforms in rat mammary gland development and carcinogenic transformation, and in mammary tumors. Female rats were injected: a) saline solution (Control mammary gland, CMG) or b) N-Nitroso-N-methyl urea (NMU), and samples were taken at 60, 90, 120 and 150 days of life. In addition, we analyzed hormone-dependent (HD) and independent (HI) tumors in ovariectomized rats, and intact tumors (IT) in non-ovariectomized ones. Our results show that, in CMG, nuclear c-Fos and proliferation decreased with age, AP-1 content was low, and nuclear ERα46/ERα66 ratio was higher than 1. In NMU, nuclear c-Fos and proliferation increased with carcinogenic transformation, AP-1 content was high, and nuclear ERα46/ERα66 was below 1. As tumor grade increased, proliferation, nuclear c-Fos and AP-1 expression were negatively associated to nuclear ERα46/ERα66 in IT. In HD, nuclear ERα46/ERα66, nuclear c-Fos expression, AP-1 levels and proliferation were lower than in HI, whose growth is estrogen-independent. Phospho-Tyr537-ERα66 content and ERK1/2 activation were associated with AP-1 levels and cell proliferation. Collectively, our findings support the notion that variant detection and ERα46/ERα66 ratio could shed light on the role of ERα isoforms in mammary gland transformation and the behavior of ERα positive mammary tumors.


Subject(s)
Estrogen Receptor alpha/genetics , Genes, fos/genetics , Mammary Neoplasms, Animal/genetics , Protein Isoforms/genetics , Animals , Cell Line, Tumor , Cell Nucleus/drug effects , Cell Nucleus/genetics , Cell Proliferation/drug effects , Cytoplasm/drug effects , Cytoplasm/genetics , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mammary Glands, Animal/metabolism , Mammary Glands, Animal/pathology , Mammary Neoplasms, Animal/metabolism , Mammary Neoplasms, Animal/pathology , Methylnitrosourea/pharmacology , Protein Isoforms/metabolism , Rats , Signal Transduction/drug effects , Transcription Factor AP-1/genetics
4.
Contrast Media Mol Imaging ; 2019: 4087895, 2019.
Article in English | MEDLINE | ID: mdl-31178670

ABSTRACT

D-α-Tocopheryl polyethylene glycol 1000 succinate (TPGS) is a Food and Drug Administration (FDA) approved biomaterial that can form nanosized micelles in aqueous solution. TPGS micelles stand as an interesting system to perform drug delivery as they can carry lipophilic drugs and overcome P glycoprotein efflux as well. Therefore, TPGS micelles combined with other copolymers have been reported in many cancer research studies as a carrier for therapeutic drugs. Their ability to reach tumoral tissue can also be exploited to develop imaging agents with diagnostic application. A radiolabeling method with 99mTc for TPGS nanosized micelles and their biodistribution in a healthy animal model as well as their pharmacokinetics and radiolabeling stability in vivo was previously reported. The aim of this work was to evaluate the performance of this radioactive probe as a diagnostic imaging agent compared to routinely available SPECT radiopharmaceutical, 99mTc-sestamibi. A small field of view gamma camera was used for scintigraphy studies using radiolabeled TPGS micelles in two animal models of breast cancer: syngeneic 4T1 murine cell line (injected in BALB/c mice) and chemically NMU-induced (Sprague-Dawley rats). Ex vivo radioactivity accumulation in organs of interest was measured by a solid scintillation counter, and a semiquantitative analysis was performed over acquired images as well. Results showed an absence of tumoral visualization in 4T1 model for both radioactive probes by gamma camera imaging. On the contrary, NMU-induced tumors had a clear tumor visualization by scintigraphy. A higher tumor/background ratio and more homogeneous uptake were found for radiolabeled TPGS micelles compared to 99mTc-sestamibi. In conclusion, 99mTc-radiolabeled TPGS micelles might be a potential SPECT imaging probe for diagnostic purposes.


Subject(s)
Breast Neoplasms/diagnostic imaging , Micelles , Nanostructures , Radiopharmaceuticals , Tomography, Emission-Computed, Single-Photon/methods , Vitamin E , Animals , Drug Evaluation, Preclinical , Drug Stability , Female , Humans , Mammary Neoplasms, Experimental/chemically induced , Mammary Neoplasms, Experimental/diagnostic imaging , Methylnitrosourea , Mice, Inbred BALB C , Mice, Inbred C3H , Radiopharmaceuticals/pharmacokinetics , Rats, Sprague-Dawley , Technetium Tc 99m Sestamibi/pharmacokinetics , Tissue Distribution , Vitamin E/pharmacokinetics
5.
Reprod Toxicol ; 69: 1-12, 2017 04.
Article in English | MEDLINE | ID: mdl-28077272

ABSTRACT

We investigated arsenite exposure on the reproductive axis of dams (during pregnancy and at cyclicity resumption) and their offspring. Pregnant rats were exposed to 5 (A5) or 50ppm (A50) of sodium arsenite in drinking water from gestational day 1 (GD1) until sacrifice at GD18 or two months postpartum. Offspring were exposed to the same treatment as their mothers from weaning to adulthood. A50-pregnant rats gained less weight, showed increased testosterone and estradiol but pregnancy was unaffected. After lactation, arsenic-exposed dams presented compromised cyclicity, decreased estradiol, increased follicle-stimulating hormone (FSH), less preovulatory follicles and presence of ovarian cysts, suggesting impaired reproduction. A50-offspring presented lower body weight; A50-female-offspring showed elevated gonadotropin releasing hormone (GnRH), FSH and testosterone, while A50-males showed diminished GnRH/FSH, but normal testosterone. We conclude that arsenite at the present exposure levels did not compromise pregnancy outcome while it negatively affected reproductive physiology in postpartum dams and their offspring.


Subject(s)
Arsenites/toxicity , Prenatal Exposure Delayed Effects , Sodium Compounds/toxicity , Adrenal Glands/drug effects , Adrenal Glands/metabolism , Animals , Arsenic/metabolism , Female , Hormones/metabolism , Hypothalamus/drug effects , Hypothalamus/metabolism , Lactation , Liver/metabolism , Male , Maternal-Fetal Exchange , Ovary/drug effects , Ovary/metabolism , Pituitary Gland/drug effects , Pituitary Gland/metabolism , Pregnancy , Rats , Rats, Sprague-Dawley , Reproduction/drug effects , Sexual Maturation/drug effects
6.
J Steroid Biochem Mol Biol ; 156: 1-9, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26518068

ABSTRACT

Endocrine disruptors (EDs) are compounds that interfere with hormone regulation and influence mammary carcinogenesis. We have previously demonstrated that the pesticide chlorpyrifos (CPF) acts as an ED in vitro, since it induces human breast cancer cells proliferation through estrogen receptor alpha (ERα) pathway. In this work, we studied the effects of CPF at environmental doses (0.01 and 1mg/kg/day) on mammary gland, steroid hormone receptors expression and serum steroid hormone levels. It was carried out using female Sprague-Dawley 40-days-old rats exposed to the pesticide during 100 days. We observed a proliferating ductal network with a higher number of ducts and alveolar structures. We also found an increased number of benign breast diseases, such as hyperplasia and adenosis. CPF enhanced progesterone receptor (PgR) along with the proliferating cell nuclear antigen (PCNA) in epithelial ductal cells. On the other hand, the pesticide reduced the expression of co-repressors of estrogen receptor activity REA and SMRT and it decreased serum estradiol (E2), progesterone (Pg) and luteinizing hormone (LH) levels. Finally, we found a persistent decrease in LH levels among ovariectomized rats exposed to CPF. Therefore, CPF alters the endocrine balance acting as an ED in vivo. These findings warn about the harmful effects that CPF exerts on mammary gland, suggesting that this compound may act as a risk factor for breast cancer.


Subject(s)
Chlorpyrifos/adverse effects , Endocrine Disruptors/adverse effects , Environmental Pollutants/adverse effects , Mammary Glands, Animal/drug effects , Mammary Glands, Animal/pathology , Pesticides/adverse effects , Animals , Estradiol/blood , Estrogen Receptor alpha/analysis , Female , Luteinizing Hormone/blood , Progesterone/analysis , Progesterone/blood , Prohibitins , Rats , Rats, Sprague-Dawley , Receptors, Progesterone/analysis
7.
Acta toxicol. argent ; 23(3): 142-152, dic. 2015. graf
Article in Spanish | LILACS | ID: biblio-837846

ABSTRACT

El clorpirifos (CPF) es un insecticida de amplio espectro que se utiliza en Argentina y en otros países de Latinoamérica. Se emplea para el control de plagas en la producción de frutas, hortalizas, cereales y plantas ornamentales. El principal mecanismo de acción descripto para este insecticida es la inhibición de la acetilcolinesterasa. Sin embargo, reportes más recientes sugieren múltiples efectos del plaguicida independientes de la inhibición de esa enzima. El objetivo de este trabajo es transmitir a la comunidad los resultados de nuestras investigaciones obtenidos utilizando diferentes dosis de CPF en distintos modelos experimentales, tanto in vitro como in vivo. En relación a esto, hemos evidenciado una acción del CPF sobre el sistema redox celular que conduce al incremento de especies reactivas del oxígeno y consecuentemente a la activación de diferentes vías de señalización. Además, hemos determinado que el insecticida CPF puede comportarse como un disruptor endócrino modulando la acción de los estrógenos y alterando la normal estructura del tejido mamario. Nuestros resultados alertan sobre el impacto que este compuesto podría tener sobre la salud, sugiriendo la necesidad de revisar su uso dado que manifiesta acciones a dosis encontradas en el ambiente.


Chlorpyrifos (CPF) is a broad spectrum insecticide used in Argentina and other Latin American countries. It is commonly used for pest control in the production of fruits, vegetables, cereals and ornamental plants. The main mechanism of action described for this insecticide is the inhibition of acetylcholinesterase activity. However, more recent reports suggest multiple effects for this pesticide in an independent way from the inhibition of this enzyme. The objective of this work is to convey to the community the results of our investigations obtained using different doses of CPF in various experimental models, both in vitro and in vivo. In this connection, we have shown a CPF action on the cellular redox system which leads to increased reactive oxygen species and the consequent activation of different signaling pathways. In addition, we have determined that the insecticide CPF acts as an endocrine disruptor modulating the action of estrogen and altering the normal structure of breast tissue. Our findings warn about the impact that this compound might have on health, suggesting the need to review its use since adverse actions were found at environmentally relevant doses.


Subject(s)
Humans , Animals , Rats , Breast Neoplasms/enzymology , Endocrine Disruptors/toxicity , Organophosphorus Compounds/toxicity , Oxidation-Reduction , Oxidative Phosphorylation , Breast Neoplasms/chemically induced , Mammary Neoplasms, Experimental , Neoplasm Metastasis/ultrastructure
8.
Chemosphere ; 120: 343-50, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25180937

ABSTRACT

It is well known the participation of oxidative stress in the induction and development of different pathologies including cancer, diabetes, neurodegeneration and respiratory disorders among others. It has been reported that oxidative stress may be induced by pesticides and it could be the cause of health alteration mediated by pollutants exposure. Large number of registered products containing chlorpyrifos (CPF) is used to control pest worldwide. We have previously reported that 50 µM CPF induces ROS generation and produces cell cycle arrest followed by cell death. The present investigation was designed to identify the pathway involved in CPF-inhibited cell proliferation in MCF-7 and MDA-MB-231 breast cancer cell lines. In addition, we determined if CPF-induced oxidative stress is related to alterations in antioxidant defense system. Finally we studied the molecular mechanisms underlying in the cell proliferation inhibition produced by the pesticide. In this study we demonstrate that CPF (50 µM) induces redox imbalance altering the antioxidant defense system in breast cancer cells. Furthermore, we found that the main mechanism involved in the inhibition of cell proliferation induced by CPF is an increment of p-ERK1/2 levels mediated by H2O2 in breast cancer cells. As PD98059 could not abolish the increment of ROS induced by CPF, we concluded that ERK1/2 phosphorylation is subsequent to ROS production induced by CPF but not the inverse.


Subject(s)
Antioxidants/metabolism , Cell Proliferation/drug effects , Chlorpyrifos/toxicity , Insecticides/toxicity , MAP Kinase Signaling System/drug effects , Oxidative Stress/drug effects , Cell Line, Tumor , Humans , MCF-7 Cells , Oxidation-Reduction , Phosphorylation/drug effects
9.
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
10.
Toxicol Lett ; 213(2): 184-93, 2012 Sep 03.
Article in English | MEDLINE | ID: mdl-22771950

ABSTRACT

It has reported that many environmental compounds may display estrogenic actions and these findings led to researchers to associate breast cancer risk with the use of some pesticides. The aim of this work was to investigate the effect of chlorpyrifos (CPF) on cell proliferation and the ERα-dependence of this action employing MCF-7 and MDA-MB-231 breast cancer cell lines. We have also analyzed CPF action on the cell cycle distribution and the cyclins that are implicated in G1-S and intra-S checkpoints. Finally, the action on cell death and ROS production were studied. We demonstrated the ability of CPF 0.05µM to induce cell proliferation through ERα in hormone-dependent breast cancer cells. In contrast, CPF 50µM induces intra-S arrest modifying checkpoints proteins, through a mechanism that may involve changes in redox balance in MCF-7. In MDA-MB-231, we have found that CPF 50µM produces an arrest in G2/M phase which could be related to the capacity of the pesticide for binding to tubulin sites altering microtubules polymerization. Altogether, our results provide new evidences on the action of the pesticide CPF as an environmental breast cancer risk factor due to the effects that causes on the mechanisms that modulate breast cell proliferation.


Subject(s)
Breast Neoplasms/chemically induced , Chlorpyrifos/toxicity , Estrogen Receptor alpha/metabolism , Insecticides/toxicity , Neoplasms, Hormone-Dependent/chemically induced , Blotting, Western , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclins/metabolism , Female , Flow Cytometry , Humans , MCF-7 Cells , Neoplasms, Hormone-Dependent/metabolism , Neoplasms, Hormone-Dependent/pathology , Oxidation-Reduction , Phosphorylation
11.
Front Biosci (Elite Ed) ; 3(3): 1042-60, 2011 06 01.
Article in English | MEDLINE | ID: mdl-21622113

ABSTRACT

In order to better understand the role of histamine H4 (H4R) receptor in breast cancer, we studied the receptor expression pattern, associated signal transduction pathway and biological responses, in breast cancer cell lines with different malignant characteristics. A different pattern of protein expression was observed in MDA-MB-231 compared to MCF-7 cells determined by western blot, exhibiting the presence of a diverse range of molecular weight species of the H4R. H4R agonist reduced cyclic adenosine monophosphate (cAMP) formation induced by forskolin only in MCF-7 cells. In MDA-MB-231 cells, H4R agonists significantly decreased cell proliferation, augmented the Annexin-V and TdT-mediated UTP-biotin Nick End labelling (TUNEL) positive cells and produced a 2.5-fold increase in cell senescence. In MCF-7 cells, H4R agonists inhibited proliferation by 50%, increasing the exponential doubling time. This effect was associated to an augment in Annexin-V and TUNEL positive cells, and a 2-fold increase in cell senescence. We conclude that H4R is functionally expressed in human breast cancer cell lines, exhibiting a key role in histamine-mediated biological processes such as cell proliferation, senescence and apoptosis.


Subject(s)
Breast Neoplasms/pathology , Cell Proliferation , Receptors, G-Protein-Coupled/physiology , Receptors, Histamine/physiology , Apoptosis , Base Sequence , Blotting, Western , Cell Line, Tumor , DNA Primers , Female , Humans , In Situ Nick-End Labeling , Receptors, Histamine H4 , Reverse Transcriptase Polymerase Chain Reaction
12.
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
13.
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
14.
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
15.
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
16.
Toxicol Sci ; 89(1): 83-92, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16237195

ABSTRACT

Hexachlorobenzene (HCB) is a widespread environmental pollutant. Controversy still exists about the breast carcinogenic properties of organochlorines in humans. The ligands, receptors, and related signaling proteins of the insulin growth factor family are involved in the regulation of breast-cancer cell growth. The aims of this study were to determine: (1) whether HCB is co-carcinogenic in a medium term assay of N-nitroso N-methylurea (NMU)-induced mammary tumors in rats; (2) the effect of HCB on insulin receptor (IR), insulin-like growth factor-I receptor (IGF-IR) and insulin receptor substrate-1 (IRS-1) levels and on IRS-1 phosphorylation; (3) microsomal and cytosolic Protein Tyrosine Kinase (PTK) activities in mammary glands and NMU-induced tumors. Sprague Dawley rats were injected with 50 mg/kg body weight of NMU at 50, 80, and 110 days old. HCB (100 mg/kg body weight) was administered three times a week from 65 to 110 days of age. Rats were separated in four groups: control, NMU, HCB, and NMU-HCB. HCB alone did not induce tumor development. Parameters of tumor development were increased in NMU-HCB compared to NMU rats. A higher cellular undifferentiation was observed in NMU-HCB tumors. IR, IGF-IR, and IRS-1 levels were higher in HCB than in controls. Conversely IGF-IR levels decreased in NMU-HCB vs. NMU group. The IRS-1 phosphorylation increased in HCB rats; however, it decreased in NMU-HCB vs. NMU. HCB decreased microsomal PTK activity in tumors. This study showed for the first time that HCB is a co-carcinogenic agent in NMU-induced mammary tumors in rats. Our results suggest that the IR and/or IGF-IR signaling pathway may be involved in the mechanism of action of HCB.


Subject(s)
Adaptor Proteins, Signal Transducing , Carcinogens/toxicity , Cocarcinogenesis , Hexachlorobenzene/toxicity , Insulin-Like Growth Factor I/metabolism , Mammary Glands, Animal/drug effects , Animals , Carcinogenicity Tests , Disease Models, Animal , Drug Therapy, Combination , Female , Hexachlorobenzene/classification , Insulin Receptor Substrate Proteins , Mammary Glands, Animal/metabolism , Mammary Neoplasms, Animal/chemically induced , Mammary Neoplasms, Animal/metabolism , Mammary Neoplasms, Animal/pathology , Methylnitrosourea , Phosphoproteins/metabolism , Protein-Tyrosine Kinases/metabolism , Rats , Rats, Sprague-Dawley , Receptor, IGF Type 1/metabolism , Receptor, Insulin/metabolism , Signal Transduction/drug effects
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 ; 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
...