Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Nora Mohamad is active.

Publication


Featured researches published by Nora Mohamad.


Cancer Biology & Therapy | 2008

The role of histamine in human mammary carcinogenesis: H3 and H4 receptors as potential therapeutic targets for breast cancer treatment.

Vanina A. Medina; Máximo Croci; Ernesto Crescenti; Nora Mohamad; Francisca Sánchez-Jiménez; Noelia A. Massari; Mariel Núñez; G. Cricco; G. Martin; Rosa Bergoc; Elena Rivera

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


Cancer Letters | 1994

Induction of mammary tumors in rat by intraperitoneal injection of NMU: histopathology and estral cycle influence

Elena Rivera; N. Andrade; G. Martin; G. Melito; G. Cricco; Nora Mohamad; Carlos Davio; Ricardo A. Caro; Rosa Bergoc

In order to obtain an experimental model we induced mammary tumors in female Sprague-Dawley rats. The carcinogen N-nitroso-N-methylurea (NMU) was injected intraperitoneally (i.p.) at doses of 50 mg/kg body weight when animals were 50, 80 and 110 days old. Tumor sizes were measured with a caliper and their growth parameters and histopathological properties were tested. For 100 rats, 88.4% of developed lesions were ductal carcinomas, histologically classified as 52.8% cribiform variety, 30.6% solid carcinoma. Metastases in liver, spleen and lung were present. Other primary tumors were detected with low incidence. The influence of the rat estrous cycle during the first exposure to intraperitoneal NMU injection was studied. The latency period in estrus, proestrus and diestrus was 82 +/- 15, 77 +/- 18 and 79 +/- 18 days, respectively. Tumor incidence was significantly higher in estrus (95.2%) than proestrus (71.4%) or diestrus (77.4), (P < 0.01). Mean number or tumors per animal was similar among the three groups (4.4 +/- 3.2, 3.8 +/- 3.6, 3.2 +/- 1.8). The procedure described appears to be the simplest method for inducing experimental mammary tumors in rats.


Cancer Biology & Therapy | 2006

Histamine-Mediated Signaling Processes in Human Malignant Mammary Cells

Vanina A. Medina; G. Cricco; Mariel Núñez; G. Martin; Nora Mohamad; F. Correa-Fiz; Francisca Sánchez-Jiménez; Rosa Bergoc; Elena Rivera

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 μM histamine reduced significantly proliferation to 23% inducing cell cycle arrest in G2/M phase, differentiation by 26% and a significant increase in the number of apoptotic cells (p


International Journal of Radiation Biology | 2007

Mechanisms underlying the radioprotective effect of histamine on small intestine.

Vanina A. Medina; Máximo Croci; Nora Mohamad; Noelia A. Massari; Gloria Garbarino; G. Cricco; Mariel Núñez; G. Martin; Ernesto Crescenti; Rosa Bergoc; Elena Rivera

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.


Inflammation Research | 2008

Histamine regulates pancreatic carcinoma cell growth through H3 and H4 receptors

G. Cricco; Nora Mohamad; Lorena Sambuco; F. Genre; Máximo Croci; Alicia Gutiérrez; Vanina A. Medina; Rosa Bergoc; Elena Rivera; G. Martin

Several lines of evidence suggest that histamine (HA) may act as an autocrine or paracrine growth factor increasing proliferation rate in normal and malignant tissues. Previously we reported that histamine H1 and H2 receptors are expressed and associated with cell growth in Panc-1, a cell line derived from a human ductal pancreatic carcinoma [1]. This work was to evaluate the presence of histamine H3 and H4 receptors and their potential involvement in PANC-1 cells proliferation.


The International Journal of Biochemistry & Cell Biology | 2014

Fibroblasts induce epithelial to mesenchymal transition in breast tumor cells which is prevented by fibroblasts treatment with histamine in high concentration

Juliana Porretti; Nora Mohamad; G. Martin; G. Cricco

Epithelial to mesenchymal transition (EMT) of cancer cells is an essential process in cancer progression. Cancer cells that undergone EMT loose cell-cell contacts, acquire mesenchymal properties and develop migratory and invasive abilities. In previous studies we have demonstrated that histamine may modify the invasive phenotype of pancreatic and mammary tumor cells. In this work we proposed to investigate whether histamine may also influence the interaction between tumor cells and normal fibroblasts. The potential activation of normal CCD-1059Sk fibroblasts by histamine and EMT phenotypic changes induced in MCF-7 and MDA-MB-231 breast tumor cells by the conditioned media (CM) derived from fibroblasts were evaluated. Initially, we determined the presence of H1, H2 and H4 histamine receptors and matrix metalloproteinase 2 (MMP2) mRNA in CCD-1059Sk fibroblasts. MMP2 gelatinolytic activity, cell migration and alpha-smooth muscle actin expression were increased in fibroblasts by low doses (<1μM) and decreased by high doses (20μM) of histamine. MCF-7 cells cultured with CM from fibroblasts exhibited spindle-shaped morphology, cell spreading and cytoplasmic expression of β-catenin but there was no change in MMP2 activity and cell migration. MDA-MB-231 cells cultured with CM from fibroblasts showed a more elongated phenotype, cell spreading, cytoplasmic β-catenin, increased MMP2 activity and endogenous TGF-β1 expression, and enhanced cell migration and invasion. Notably, all these features were reversed when mammary tumor cells were cultured with CM from fibroblasts treated with 20μM histamine. In conclusion, high doses of histamine may prevent the activation of fibroblasts and also avert the EMT related changes induced in epithelial tumor cells by fibroblasts CM.


Inflammation Research | 2006

Histamine modulates cellular events involved in tumour invasiveness in pancreatic carcinoma cells.

G. Cricco; Mariel Núñez; Vanina A. Medina; Gloria Garbarino; Nora Mohamad; Alicia Gutiérrez; Claudia Cocca; Rosa Bergoc; Elena Rivera; G. Martin

No Abstract..


Inflammation Research | 2005

Histamine is a selective protector against cellular damage produced by ionizing radiation

Vanina A. Medina; G. Cricco; Nora Mohamad; Máximo Croci; Mariel Núñez; G. Martin; Claudia Cocca; Rosa Bergoc; Elena Rivera

No Abstract..


Pharmacological Research | 2016

Histamine prevents radiation-induced mesenchymal changes in breast cancer cells.

Tamara E. Galarza; Nora Mohamad; Mónica A. Táquez Delgado; Guadalupe M. Vedoya; Ernesto Crescenti; Rosa Bergoc; G. Martin; G. Cricco

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


Archive | 2011

Histamine and Breast Cancer: A New Role for a Well Known Amine

G. Cricco; Nora Mohamad; María Soledad Sáez; Eduardo Valli; Elena Rivera; G. Martin

Metastases are the most devastating aspect of cancer since most deaths from cancer are related to them. The ability of tumors to invade the neighboring extracellular matrix, which is primarily accompanied by augmented matrix metalloproteinases (MMPs) production and cell migration, is critical for metastases. After surgical removal of primary breast tumors, malignant cells may still remain and radiotherapy is an efficient modality to reduce the risk of local recurrence. However, proliferative, invasive, and metastatic capacities can be increased in the surviving tumor cells of irradiated breast and other neoplasias (Baluna et al, 2006; Tsukamoto et al, 2007; Tsutsumi et al, 2009). To improve the efficacy of radiotherapy, this phenomenon must be further studied to elaborate therapeutic modalities to prevent radiation enhancement of cancer cell invasion. Histamine is an endogenous biogenic amine extensively distributed throughout the organism which exerts multiple functions in physiologic and pathophysiological processes by stimulation of four G-protein coupled receptors (H1, H2, H3 and H4 histamine receptors) with different tissue expression patterns and functions. It is well known that diverse tumoral tissues and cell lines express the different histamine receptors through which histamine brings about its effects on cell proliferation, differentiation, survival and death. A great deal of evidence shows a relevant role of histamine in tumor progression, however controversial results are published depending on the cell type and the histamine receptor subtype that is activated (Blaya et al, 2010; Francis et al, 2009; Parsons & Ganellin, 2006; Soule et al, 2010). It has also been determined that numerous tumour tissues and cell lines express L-histidine decarboxylase, the histamine-synthesizing enzyme, and contain high levels of endogenous histamine which released to the extracellular media may exert its effects via a paracrine or autocrine regulation (Falus et al, 2001; Pos et al, 2004; Rivera et al, 2000). Additionally some effects on tumor growth may be mediated by histamine regulation of angiogenesis and immunity (Lazar-Molnar et al, 2002; Tomita et al, 2003). Our research team has demonstrated the expression of histamine membrane receptors and their association to different signalling pathways in breast cancer biopsies and a large number of transformed cell lines, being our works the first ones to report the presence of H3

Collaboration


Dive into the Nora Mohamad's collaboration.

Top Co-Authors

Avatar

G. Cricco

University of Buenos Aires

View shared research outputs
Top Co-Authors

Avatar

G. Martin

University of Buenos Aires

View shared research outputs
Top Co-Authors

Avatar

Elena Rivera

University of Buenos Aires

View shared research outputs
Top Co-Authors

Avatar

Rosa Bergoc

University of Buenos Aires

View shared research outputs
Top Co-Authors

Avatar

Vanina A. Medina

University of Buenos Aires

View shared research outputs
Top Co-Authors

Avatar

Mariel Núñez

University of Buenos Aires

View shared research outputs
Top Co-Authors

Avatar

Alicia Gutiérrez

University of Buenos Aires

View shared research outputs
Top Co-Authors

Avatar

Máximo Croci

University of Buenos Aires

View shared research outputs
Top Co-Authors

Avatar

Claudia Cocca

University of Buenos Aires

View shared research outputs
Top Co-Authors

Avatar

Ernesto Crescenti

University of Buenos Aires

View shared research outputs
Researchain Logo
Decentralizing Knowledge