Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
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
2.
Front Biosci (Schol Ed) ; 7(1): 1-9, 2015 06 01.
Article in English | MEDLINE | ID: mdl-25961682

ABSTRACT

Breast cancer is the second most common cancer worldwide, and the leading cause of cancer death in women. Several studies underlined the critical role of histamine in breast cancer development and progression. This review addresses the latest evidence regarding the involvement of histamine and histamine receptors in breast cancer, focusing particularly in the histamine H4 receptor (H4R). Histamine concentration in breast cancer tissues was found to be higher than that in normal tissues of healthy controls by means of an increase in the activity of histidine decarboxylase (HDC), the enzyme involved in histamine production. The expression of H4R in different experimental models and human biopsies, the associated biological responses, as well as the in vivo treatment of experimental tumors with H4R ligands is reviewed. Evidence demonstrates that the H4R exhibits a key role in histamine-mediated biological processes such as cell proliferation, senescence and apoptosis in breast cancer. The polymorphisms of the H4R and HDC genes and their association with breast cancer risk and malignancy reinforce the critical (patho)physiological role of H4R in breast cancer. In addition, H4R agonists display anti-tumor effects in vivo in a triple negative breast cancer model. The findings support the exploitation of the H4R as a molecular target for breast cancer drug development.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/antagonists & inhibitors , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Female , Heterografts , Histamine/metabolism , Histamine Antagonists/therapeutic use , Humans , Ligands , Molecular Targeted Therapy , Receptors, G-Protein-Coupled/metabolism , Receptors, Histamine/metabolism , Receptors, Histamine H4
3.
Cancer Biol Ther ; 16(1): 137-48, 2015.
Article in English | MEDLINE | ID: mdl-25482934

ABSTRACT

The radioprotective potential of histamine on healthy tissue has been previously demonstrated. The aims of this work were to investigate the combinatorial effect of histamine or its receptor ligands and gamma radiation in vitro on the radiobiological response of 2 breast cancer cell lines (MDA-MB-231 and MCF-7), to explore the potential molecular mechanisms of the radiosensitizing action and to evaluate the histamine-induced radiosensitization in vivo in a triple negative breast cancer model. Results indicate that histamine significantly increased the radiosensitivity of MDA-MB-231 and MCF-7 cells. This effect was mimicked by the H1R agonist 2-(3-(trifluoromethyl)phenyl)histamine and the H4R agonists (Clobenpropit and VUF8430) in MDA-MB-231 and MCF-7 cells, respectively. Histamine and its agonists enhanced radiation-induced oxidative DNA damage, DNA double-strand breaks, apoptosis and senescence. These effects were associated with increased production of reactive oxygen species, which correlated with the inhibition of catalase, glutathione peroxidase and superoxide dismutase activities in MDA-MB-231 cells. Histamine was able also to potentiate in vivo the anti-tumoral effect of radiation, increasing the exponential tumor doubling time. We conclude that histamine increased radiation response of breast cancer cells, suggesting that it could be used as a potential adjuvant to enhance the efficacy of radiotherapy.


Subject(s)
Breast Neoplasms/metabolism , Histamine/metabolism , Radiation Tolerance , Radiation, Ionizing , Animals , Antioxidants/metabolism , Apoptosis/drug effects , Apoptosis/radiation effects , Breast Neoplasms/pathology , Breast Neoplasms/radiotherapy , Cell Line, Tumor , Cellular Senescence/drug effects , Cellular Senescence/radiation effects , DNA Damage/drug effects , DNA Damage/radiation effects , Disease Models, Animal , Dose-Response Relationship, Drug , Dose-Response Relationship, Radiation , Female , Histamine/pharmacology , Humans , MCF-7 Cells , Oxidation-Reduction , Radiation Tolerance/drug effects , Radiation-Sensitizing Agents/pharmacology , Reactive Oxygen Species/metabolism , Tumor Burden/drug effects , Tumor Burden/radiation effects , Xenograft Model Antitumor Assays
4.
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
5.
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
6.
PLoS One ; 8(7): e69106, 2013.
Article in English | MEDLINE | ID: mdl-23922686

ABSTRACT

Based on previous data on the histamine radioprotective effect on highly radiosensitive tissues, in the present work we aimed at investigating the radioprotective potential of the H4R ligand, JNJ7777120, on ionizing radiation-induced injury and genotoxic damage in small intestine, salivary glands and hematopoietic tissue. For that purpose, rats were divided into 4 groups. JNJ7777120 and JNJ7777120-irradiated groups received a daily subcutaneous JNJ7777120 injection (10 mg/kg) starting 24 h before irradiation. Irradiated groups received a single dose of 5 Gy on whole-body using Cesium-137 source and were sacrificed 3 or 30 days after irradiation. Tissues were removed, fixed, stained with hematoxylin and eosin or PAS staining and histological characteristics were evaluated. Proliferative and apoptotic markers were studied by immunohistochemistry, while micronucleus assay was performed to evaluate DNA damage. Submandibular gland (SMG) function was evaluated by methacholine-induced salivation. Results indicate that JNJ7777120 treatment diminished mucosal atrophy and preserved villi and the number of crypts after radiation exposure (240±8 vs. 165±10, P<0.01). This effect was associated to a reduced apoptosis and DNA damage in intestinal crypts. JNJ7777120 reduced radiation-induced aplasia, preserving medullar components and reducing formation of micronucleus and also it accelerated bone marrow repopulation. Furthermore, it reduced micronucleus frequency in peripheral blood (27±8 vs. 149±22, in 1,000 erythrocytes, P<0.01). JNJ7777120 completely reversed radiation-induced reduced salivation, conserving glandular mass with normal histological appearance and reducing apoptosis and atrophy of SMG. JNJ7777120 exhibits radioprotective effects against radiation-induced cytotoxic and genotoxic damages in small intestine, SMG and hematopoietic tissues and, thus, could be of clinical value for patients undergoing radiotherapy.


Subject(s)
Hematopoietic System/drug effects , Indoles/pharmacology , Intestine, Small/drug effects , Piperazines/pharmacology , Radiation-Protective Agents/pharmacology , Receptors, G-Protein-Coupled/metabolism , Receptors, Histamine/metabolism , Salivary Glands/drug effects , Animals , Bone Marrow/drug effects , Bone Marrow/pathology , Bone Marrow/radiation effects , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Hematopoietic System/pathology , Hematopoietic System/radiation effects , Humans , Intestine, Small/pathology , Intestine, Small/radiation effects , Ligands , Male , Mutagens/toxicity , Rats , Rats, Sprague-Dawley , Receptors, Histamine H4 , Salivary Glands/pathology , Salivary Glands/radiation effects , Sulfhydryl Compounds/metabolism , Thiobarbituric Acid Reactive Substances/metabolism , Whole-Body Irradiation
7.
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
8.
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
9.
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
10.
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
11.
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
12.
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
13.
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
14.
Br J Pharmacol ; 161(4): 755-67, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20636392

ABSTRACT

Considerable evidence has been collected indicating that histamine can modulate proliferation of different normal and malignant cells. High histamine biosynthesis and content together with histamine receptors have been reported in different human neoplasias including melanoma, colon and breast cancer, as well as in experimental tumours in which histamine has been postulated to behave as an important paracrine and autocrine regulator of proliferation. The discovery of the human histamine H(4) receptor in different tissues has contributed to our understanding of histamine role in numerous physiological and pathological conditions revealing novel functions for histamine and opening new perspectives in histamine pharmacology research. In the present review we aimed to briefly summarize current knowledge on histamine and histamine receptor involvement in cancer before focusing on some recent evidence supporting the novel role of histamine H(4) receptor in cancer progression representing a promising molecular target and avenue for cancer drug development.


Subject(s)
Drug Delivery Systems , Neoplasms/physiopathology , Receptors, Histamine/metabolism , Animals , Antineoplastic Agents/pharmacology , Cell Proliferation , Drug Design , Histamine/metabolism , Humans , Neoplasms/drug therapy , Receptors, G-Protein-Coupled/drug effects , Receptors, G-Protein-Coupled/metabolism , Receptors, Histamine/drug effects , Receptors, Histamine H4
15.
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
16.
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
17.
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
18.
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
19.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...