Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Br J Pharmacol ; 170(1): 188-99, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23425150

ABSTRACT

BACKGROUND AND PURPOSE: The presence of the histamine H4 receptor (H4R) was previously reported in benign and malignant lesions and cell lines derived from the human mammary gland. The aim of this work was to evaluate the effects of H4R ligands on the survival, tumour growth rate and metastatic capacity of breast cancer in an experimental model. EXPERIMENTAL APPROACH: Xenograft tumours of the highly invasive human breast cancer cell line MDA-MB-231 were established in immune deficient nude mice. The following H4R agonists were employed: histamine (5 mg kg⁻¹), clozapine (1 mg kg⁻¹) and the experimental compound JNJ28610244 (10 mg kg⁻¹). RESULTS: Data indicate that developed tumours were highly undifferentiated, expressed H4R and exhibited high levels of histamine content and proliferation marker (PCNA) while displaying low apoptosis. Mice of the untreated group displayed a median survival of 60 days and a tumour doubling time of 7.4 ± 0.6 days. A significant decrease in tumour growth evidenced by an augment of the tumour doubling time was observed in the H4R agonist groups (13.1 ± 1.2, P < 0.01 in histamine group; 15.1 ± 1.1, P < 0.001 in clozapine group; 10.8 ± 0.7, P < 0.01 in JNJ28610244 group). This effect was associated with a decrease in the PCNA expression levels, and also reduced intratumoural vessels in histamine and clozapine treated mice. Histamine significantly increased median survival (78 days; Log rank Mantel-Cox Test, P = 0.0025; Gehan-Breslow-Wilcoxon Test, P = 0.0158) and tumoural apoptosis. CONCLUSIONS AND IMPLICATIONS: Histamine through the H4R exhibits a crucial role in tumour progression. Therefore, H4R ligands offer a novel therapeutic potential as adjuvants for breast cancer treatment.


Subject(s)
Breast Neoplasms/drug therapy , Histamine Agonists/pharmacology , Histamine/metabolism , Receptors, G-Protein-Coupled/agonists , Animals , Apoptosis/drug effects , Breast Neoplasms/pathology , Cell Line, Tumor , Clozapine/pharmacology , Disease Progression , Female , Gene Expression Regulation, Neoplastic , Histamine/pharmacology , Humans , Indoles/pharmacology , Mice , Mice, Nude , Oximes/pharmacology , Proliferating Cell Nuclear Antigen/genetics , Receptors, G-Protein-Coupled/metabolism , Receptors, Histamine/metabolism , Receptors, Histamine H4 , Survival Rate , Xenograft Model Antitumor Assays
2.
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
3.
Inflamm Res ; 61(5): 455-64, 2012 May.
Article in English | MEDLINE | ID: mdl-22261710

ABSTRACT

OBJECTIVE: We have recently reported that experimental periodontitis (EP) reduced methacholine-induced submandibular gland (SMG) salivary secretion. The aim of the present study was to determine whether histamine could prevent SMG impairment produced by EP. MATERIALS AND METHODS: Bilateral EP was induced for 2 weeks and histamine treatment (0.1 mg/kg subcutaneously) was started 5 days before the end of the experimental period in male rats. The histamine effects on periodontitis-altered functional and histological parameters of SMG and on periodontal bone loss were evaluated. RESULTS: Histamine treatment partially reversed the methacholine-induced salivation reduction produced by EP while preventing SMG histological damage. Histamine's effect on SMG was associated with an increased proliferation rate (2.2 ± 0.3 vs. 0.2 ± 0.2 proliferative cells per field, P < 0.001). Furthermore, histamine completely prevented enhanced EP-induced apoptosis (1.0 ± 0.4 vs. 60.9 ± 4.6 apoptotic cells per field, P < 0.001). The protective effect exerted by histamine on SMG functionality is associated with attenuation of lingual and vestibular bone loss (0.66 ± 0.04 vs. 0.97 ± 0.06 mm; P < 0.001). CONCLUSIONS: Histamine is able to reduce periodontitis-induced damage to SMG and bone structure.


Subject(s)
Histamine/therapeutic use , Periodontitis/drug therapy , Salivation/drug effects , Submandibular Gland/drug effects , Alveolar Bone Loss/prevention & control , Animals , Apoptosis/drug effects , Cell Proliferation/drug effects , Disease Progression , Histamine/pharmacology , Male , Periodontal Diseases , Periodontitis/pathology , Periodontitis/physiopathology , Rats , Submandibular Gland/pathology , Submandibular Gland/physiology
4.
J Radiat Res ; 52(5): 557-67, 2011.
Article in English | MEDLINE | ID: mdl-21952314

ABSTRACT

In this study we first evaluated the general radioprotective efficacy of Se, Zn and Mn (4 µg/ml each) plus Lachesis muta venom (4 ng/ml) combination (O-LM) by determining survival on rats irradiated with lethal doses of gamma-rays. The aim of the second part of the study was to investigate the O-LM ability to prevent ionizing radiation-induced damage on small intestine, bone marrow and submandibular glands. Hence, histological characteristics and functional studies, together with proliferation and apoptotic marker levels on whole body irradiated rats with a 5 Gy dose were evaluated. Results show that all animals of the untreated group died after whole body irradiation with 8 and 10 Gy while 60 day-survival was more than 80% and 40% in O-LM-treated animals, respectively. Histopathological examinations revealed a high degree of small intestine and submandibular gland radioprotection 3 days post-irradiation. O-LM inhibited histological damage on small intestine, restoring the radiation-induced reduction in villous height and crypt number. O-LM prevented radiation-induced loss of salivary gland function and morphological alterations. These effects were associated to a complete inhibition of radiation-induced apoptosis. Furthermore, studies performed 30 days post-irradiation revealed that O-LM significantly improved bone marrow repopulation, increasing all medullar progenies to the extent of the non-irradiated animals, and completely prevented permanent submandibular gland alterations. Based on the present results and taking into account that O-LM is being safely administered in phase I clinical trial as an immunomodulator, we conclude that O-LM is a non-toxic promising approach to achieve radioprotection for patients undergoing radiotherapy.


Subject(s)
Radiation-Protective Agents/pharmacology , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Bone Marrow/drug effects , Bone Marrow/injuries , Bone Marrow/radiation effects , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Crotalid Venoms/administration & dosage , Crotalid Venoms/pharmacology , Gamma Rays/adverse effects , Humans , Intestine, Small/drug effects , Intestine, Small/injuries , Intestine, Small/radiation effects , Male , Manganese/administration & dosage , Manganese/pharmacology , Radiation Injuries, Experimental/pathology , Radiation Injuries, Experimental/prevention & control , Radiation-Protective Agents/administration & dosage , Rats , Rats, Sprague-Dawley , Selenium/administration & dosage , Selenium/pharmacology , Submandibular Gland/drug effects , Submandibular Gland/injuries , Submandibular Gland/radiation effects , Zinc/administration & dosage , Zinc/pharmacology
5.
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
6.
Int J Radiat Biol ; 87(3): 284-92, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21142703

ABSTRACT

PURPOSE: Xerostomia is a common, disturbing side-effect among patients treated with radiotherapy for head-and-neck cancer. The aim of the present work was to investigate whether histamine could prevent salivary gland dysfunction and histological alterations exerted by ionising radiation. MATERIALS AND METHODS: Forty-eight rats were divided into four groups. Histamine and histamine-5 Gy groups received a daily subcutaneous histamine injection (0.1 mg/kg) starting 24 h before irradiation. Histamine-5 Gy and untreated-5 Gy groups were irradiated with a single dose of whole-body Cesium-137 irradiation. Control and untreated-5 Gy groups were given daily saline injections. Three days post irradiation metacholine-induced salivary secretion was measured or animals were sacrificed and submandibular gland (SMG) removed, stained and histological characteristics were evaluated. Proliferation and apoptosis markers were studied by immunohistochemistry. RESULTS: Radiation decreased salivary secretion by 40% in comparison to untreated rats, which was associated with loss of SMG mass, alteration of epithelial architecture, partial loss of secretor granular material, diminished proliferation and a remarkable apoptotic response. In contrast, histamine completely reversed the reduced salivation induced by radiation, conserved glandular mass with normal appearance and preserved the structural organisation of secretor granules. Radiation-induced toxicity is prevented by histamine essentially by suppressing apoptosis of ductal and acinar cells, reducing the number of apoptotic cells per field (19.0 ± 3.8 vs. 106.0 ± 12.0 in untreated animals, P < 0.001), and also by preventing the radiation-induced decrease in cell proliferation. CONCLUSIONS: Histamine prevents morphological and functional radiation-induced damage on SMG, representing a potential radioprotector for treatment of patients undergoing radiotherapy for head and neck malignancies.


Subject(s)
Histamine/metabolism , Submandibular Gland/drug effects , Submandibular Gland/radiation effects , Xerostomia/etiology , Animals , Apoptosis , Head and Neck Neoplasms/radiotherapy , Male , Radiation Injuries/pathology , Radiation, Ionizing , Radiotherapy/adverse effects , Rats , Rats, Sprague-Dawley , Salivary Glands/drug effects , Salivary Glands/radiation effects , Xerostomia/prevention & control
7.
Vaccine ; 29(4): 728-36, 2011 Jan 17.
Article in English | MEDLINE | ID: mdl-21095252

ABSTRACT

We investigated the use of a live, attenuated Salmonella enterica serovar Typhi vaccine strain as an antitumor immunotherapy. Mice bearing a subcutaneous tumor (LM3 mammary adenocarcinoma) were immunized on three occasions with S. Typhi strain CVD 915 by injection into the tumor, the peritumoral tissue and the draining lymph node areas; this procedure was termed Salmonella multiple treatment (Salmonella MT). Tumor-bearing mice subjected to the Salmonella MT exhibited reduced tumor growth, prolonged survival and reduced incidence of lung metastases, compared to untreated mice. We examined the mechanisms mediating this effect and found that Salmonella MT promoted an antitumor Th1-type response characterized by increased frequencies of IFN-γ-secreting CD4(+) T and CD8(+) T cells with reduction of regulatory T cells in tumor draining lymph nodes. The main cells infiltrating bacteria-treated tumors were activated neutrophils, which can exert an antitumor effect through the secretion of TNF-α. These results demonstrate for the first time the efficacy of an attenuated S. Typhi vaccine strain as a cancer immunotherapeutic agent. By potentiating the host antitumor immune response, this approach could be a powerful adjunct tool for cancer therapy.


Subject(s)
Adenocarcinoma/therapy , Breast Neoplasms/therapy , Cancer Vaccines/immunology , Immunity, Cellular , Immunotherapy/methods , Neutrophil Activation , Salmonella typhi/immunology , Adenocarcinoma/pathology , Animals , Breast Neoplasms/pathology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/administration & dosage , Disease Models, Animal , Female , Mice , Mice, Inbred BALB C , Neoplasm Metastasis/prevention & control , Survival Analysis , Th1 Cells/immunology , Tumor Necrosis Factor-alpha/metabolism , Vaccination/methods , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/immunology
8.
Int J Radiat Biol ; 86(4): 283-90, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20353338

ABSTRACT

PURPOSE: Based on our previous data on the histamine radioprotective effect on small intestine, in the present work we aimed to determine whether histamine is able to protect bone marrow cells against ionising radiation damage. MATERIALS AND METHODS: 56 mice and 40 rats were divided into four groups. Histamine and histamine-irradiated groups received a daily subcutaneous histamine injection (0.1 mg/kg) starting 24 h before irradiation. Irradiated groups received a single dose on whole-body using Cesium-137 source and were sacrificed three days after irradiation. We evaluated the number of medullar components, bone marrow trophism, oedema, vascular damage, and other histological characteristics and also proliferation markers by immunohistochemistry. RESULTS: Histamine treatment substantially reduced the grade of aplasia, the oedema and vascular damage induced by ionising radiation on bone marrow of mice and rats. Additionally, histamine preserved medullar components increasing the number of megakaryocytes (14.0 +/- 1.0 vs. 7.3 +/- 1.0 in mice; and 9.9 +/- 1.3 vs. 4.1 +/- 1.0 in rats, P < 0.01) and also myeloid (253.4 +/- 37.6 vs. 7.8 +/- 1.5 in mice; and 52.0 +/- 3.7 vs. 31.8 +/- 3.1 in rats, P < 0.01), lymphoid (97.4 +/- 6.5 vs. 19.8 +/- 1.6 in mice; and 23.4 +/- 0.9 vs. 11.7 +/- 2.5 in rats, P < 0.01) and erythroid cells (165.0 +/- 9.1 vs. 8.8 +/- 2.8 in mice; and 27.3 +/- 2.3 vs. 15.6 +/- 3.5 in rats, P < 0.01) per mm(2). This effect was associated with an increased proliferation rate of bone marrow cells. CONCLUSIONS: Histamine reduces ionising radiation toxicity on bone marrow cells being a suitable candidate for use as radioprotector, especially for patients undergoing radiotherapy who are at the risk of bone marrow or small intestine damage.


Subject(s)
Bone Marrow Cells/drug effects , Bone Marrow Cells/radiation effects , Bone Marrow/pathology , Histamine/pharmacology , Megakaryocytes/drug effects , Radiation-Protective Agents/pharmacology , Animals , Bone Marrow/drug effects , Bone Marrow/metabolism , Bone Marrow/radiation effects , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Cesium Isotopes , Male , Megakaryocytes/cytology , Megakaryocytes/metabolism , Megakaryocytes/radiation effects , Mice , Mice, Nude , Rats , Rats, Sprague-Dawley , Time Factors , Whole-Body Irradiation
9.
J Radiat Res ; 50(6): 537-44, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19823008

ABSTRACT

In this study we evaluated in vivo the tolerance induced by the combination of Se, Zn and Mn (4 microg/ml each) plus Lachesis muta venom (4 ng/ml) (O-LM) to high doses of ionizing radiation. The protective effect of O-LM was investigated on the small-intestine and bone marrow of mice irradiated with a single whole-body dose of 10 Gy employing a (137)Cs source. Mice were sacrificed 3 days after irradiation. Mice receiving a subcutaneous daily O-LM injection starting 30 days before irradiation, showed a higher number of crypts, enhanced villous conservation and lack of edema or vascular damage in comparison to the untreated and irradiated group. In addition, O-LM treatment decreased vascular damage and the grade of aplasia preserving medullar progenies induced by ionizing radiation on mouse bone marrow. The protective effect of O-LM against radiation injury to the small intestine was associated with an increase in proliferation and a reduction of apoptosis in intestinal crypts and furthermore, to an enhanced intestinal immunoreactivity of MnSOD, and CuZnSOD, and also catalase. Based on the present results and taking into account that O-LM is being safely administered in phase I clinical trial as an immunomodulator, we suggest that O-LM could be an attractive candidate as a safe radioprotective agent for patients undergoing radiotherapy.


Subject(s)
Bone Marrow/drug effects , Bone Marrow/pathology , Bone Marrow/radiation effects , Crotalid Venoms/administration & dosage , Intestine, Small/drug effects , Intestine, Small/radiation effects , Metals, Heavy/administration & dosage , Whole-Body Irradiation/adverse effects , Animals , Drug Combinations , Intestine, Small/pathology , Mice , Mice, Nude , Radiation Tolerance/drug effects , Radiation-Protective Agents/administration & dosage
10.
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
11.
Nucl Med Biol ; 35(2): 233-7, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18312834

ABSTRACT

OBJECTIVE: The objective of this study was to design and evaluate a 32P patch for the treatment of skin diseases. MATERIALS AND METHODS: The patch was prepared from chromic phosphate 32P and silicone. Bioelimination and biodistribution in healthy and treated animals, and the therapeutic efficacy of two treatment schemes (single dose and fractionated dose) in an animal model of skin cancer were studied. RESULTS: Based on the bioelimination and biodistribution studies, no leakage of 32P from the patch was observed. The treated tumors reduced their mean diameter compared to controls. The single-dose therapeutic scheme showed a higher number of complete and partial remissions compared to the fractionated scheme. These results were confirmed by histopathological analysis of the samples. CONCLUSION: The 32P patch was designed and produced according to specifications for the treatment of superficial lesions of the skin. Although the 32P patch is an open source, it behaves like a sealed one for use in brachytherapy treatments.


Subject(s)
Brachytherapy/instrumentation , Phosphorus Radioisotopes/pharmacokinetics , Skin Neoplasms/radiotherapy , Administration, Cutaneous , Animals , Brachytherapy/methods , Disease Models, Animal , Dose Fractionation, Radiation , Dose-Response Relationship, Radiation , Female , Metabolic Clearance Rate , Mice , Phosphorus Radioisotopes/therapeutic use , Tissue Distribution , Treatment Outcome
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.
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
15.
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
16.
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
17.
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
18.
Oncol Res ; 15(6): 301-11, 2005.
Article in English | MEDLINE | ID: mdl-16408695

ABSTRACT

The objective of this study was to evaluate the antitumor effect of glibenclamide (Gli) alone or in combination with tamoxifen (Tam) on experimental mammary tumors induced by N-nitroso-N-methylurea (NMU) in nondiabetic and diabetic rats. For experimental diabetes induction, Sprague-Dawley rats were injected with streptozotocin (STZ) on the second day of life. For experimental mammary tumor induction, nondiabetic and diabetic rats were injected IP with NMU at 50, 80, and 110 days of life. Nondiabetic and diabetic rats bearing mammary tumors were treated with 0.06 mg/day of Gli orally, Tam 1 mg/kg/day SC, or with the combined treatment (Gli + Tam). After 20 days of treatment, different responses were observed. In nondiabetic rats, 64% of tumors were responsive to Gli treatment (they regressed or remained stable), whereas 57% of tumors under treatment with Tam exhibited a response. Results of the combined Gli + Tam treatment indicated that all tumors were responsive: 58% regressed and 42% remained stable. Diabetic rats receiving Gli treatment did not show response to this treatment, while 65% of the tumors of Tam-treated diabetic rats showed regression. Histopathologic observation indicated an important intratumor secretion in all tumors of Gli-, Tam-, or Gli + Tam-treated rats. No secondary toxic effect was observed after treatment at any assayed doses. In conclusion, the present data demonstrate the in vivo antitumor action of Gli treatment on the experimental mammary tumors employed, indicating that Gli exerted a direct effect on tumor cells in nondiabetic rats. The combined Gli + Tam treatment potentiated the antitumor effect of each drug alone. Future research will examine the molecular aspects of these findings.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Diabetes Mellitus, Experimental/complications , Glyburide/pharmacology , Hypoglycemic Agents/pharmacology , Mammary Neoplasms, Experimental/drug therapy , Animals , Apoptosis/drug effects , Blood Glucose/metabolism , Diabetes Mellitus, Experimental/blood , Drug Interactions , Female , Glucose Tolerance Test , Glyburide/administration & dosage , Hypoglycemic Agents/administration & dosage , Insulin/blood , Mammary Neoplasms, Experimental/blood , Mammary Neoplasms, Experimental/complications , Mammary Neoplasms, Experimental/pathology , Methylnitrosourea , Rats , Rats, Sprague-Dawley , Tamoxifen/administration & dosage
19.
Cancer Detect Prev ; 27(1): 37-46, 2003.
Article in English | MEDLINE | ID: mdl-12600416

ABSTRACT

The aim of this study was to compare mammary gland tumorigenesis in diabetic and non-diabetic rats. Streptozotocin and N-nitroso-N-methylurea were used to induce diabetes and mammary tumors, respectively. A suppression of mammary carcinogenesis in diabetic rats was shown by a longer latency period, a lower number of tumors per animal and a smaller final tumor volume. An 84% of the lesions developed in diabetic animals were benign tumors. Eighty day-old diabetic rats had significantly lower plasma levels of total-IGF-I and insulin versus non-diabetic rats. We postulate that the decrease in the total IGF-I and insulin levels during the promotion phase of carcinogenesis in this model plays an important role in retarding the tumor development in diabetic animals and in favoring the development of benign mammary lesions.


Subject(s)
Diabetes Mellitus, Experimental/complications , Mammary Neoplasms, Experimental/complications , Mammary Neoplasms, Experimental/pathology , Alkylating Agents/toxicity , Animals , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/pathology , Female , Genes, fos/physiology , Immunohistochemistry , Insulin/blood , Insulin-Like Growth Factor I/analysis , Mammary Neoplasms, Experimental/blood , Mammary Neoplasms, Experimental/metabolism , Methylnitrosourea/toxicity , Pancreas/pathology , Proliferating Cell Nuclear Antigen/biosynthesis , Rats , Time Factors
20.
ScientificWorldJournal ; 2: 1426-32, 2002 May 22.
Article in English | MEDLINE | ID: mdl-12805928

ABSTRACT

Histopathological effects of cAMP analog (8-Chloro-cAMP), tamoxifen, and medroxyprogesterone, alone or combined, upon BALB/c mice uteri are reported. 8-Chloro-cAMP diminished uterine weight, but did not modify its histopathology or estral cycle significantly. Tamoxifen diminished uterine weight showing cystic hyperplasia and an estral cycle arrested at diestrus. Medroxyprogesterone increased uterine weight, caused a swelling of the endometrium and a pseudopregnancy estrus. When combined with 8-Chloro-cAMP, tamoxifen or medroxyprogesterone always had a predominant effect. We concluded that the effects of 8-Chloro-cAMP on mice uteri did not cause significant changes on its histopathology, but diminished its weight.


Subject(s)
8-Bromo Cyclic Adenosine Monophosphate/analogs & derivatives , 8-Bromo Cyclic Adenosine Monophosphate/adverse effects , Tamoxifen/adverse effects , Uterus/drug effects , Animals , Endometrial Hyperplasia/chemically induced , Endometrium/drug effects , Endometrium/pathology , Estrus/drug effects , Female , Medroxyprogesterone/adverse effects , Mice , Mice, Inbred BALB C , Organ Size/drug effects , Pregnancy , Pseudopregnancy/chemically induced , Uterus/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...