Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Antioxidants (Basel) ; 11(10)2022 Oct 21.
Article in English | MEDLINE | ID: mdl-36290800

ABSTRACT

Head and neck squamous cell carcinoma (HNSCC) is a remarkably heterogeneous disease with around 50% mortality, a fact that has prompted researchers to try new approaches to improve patient survival. Hemoxygenase-1 (HO-1) is the rate-limiting step for heme degradation into carbon monoxide, free iron and biliverdin. We have previously reported that HO-1 protein is upregulated in human HNSCC samples and that it is localized in the cytoplasmic and nuclear compartments; additionally, we have demonstrated that HO-1 nuclear localization is associated with malignant progression. In this work, by using pharmacological and genetic experimental approaches, we begin to elucidate the mechanisms through which HO-1 plays a role in HNSCC. We found that high HO-1 mRNA was associated with decreased patient survival in early stages of HNSCC. In vitro experiments have shown that full-length HO-1 localizes in the cytoplasm, and that, depending on its enzymatic activity, it increases cell viability and promotes cell cycle progression. Instead, HO-1 does not alter migration capacity. Furthermore, we show that C-terminal truncated HO-1 localizes into the nucleus, increases cell viability and promotes cell cycle progression. In conclusion, we herein demonstrate that HO-1 displays protumor activities in HNSCC that depend, at least in part, on the nuclear localization of HO-1.

2.
Antioxidants (Basel) ; 10(1)2021 Jan 11.
Article in English | MEDLINE | ID: mdl-33440611

ABSTRACT

Heme Oxygenase-1 (HO-1) is a type II detoxifying enzyme that catalyzes the rate-limiting step in heme degradation leading to the formation of equimolar quantities of carbon monoxide (CO), free iron and biliverdin. HO-1 was originally shown to localize at the smooth endoplasmic reticulum membrane (sER), although increasing evidence demonstrates that the protein translocates to other subcellular compartments including the nucleus. The nuclear translocation occurs after proteolytic cleavage by proteases including signal peptide peptidase and some cysteine proteases. In addition, nuclear translocation has been demonstrated to be involved in several cellular processes leading to cancer progression, including induction of resistance to therapy and enhanced metastatic activity. In this review, we focus on nuclear HO-1 implication in pathophysiological conditions with special emphasis on malignant processes. We provide a brief background on the current understanding of the mechanisms underlying how HO-1 leaves the sER membrane and migrates to the nucleus, the circumstances under which it does so and, maybe the most important and unknown aspect, what the function of HO-1 in the nucleus is.

3.
J Steroid Biochem Mol Biol ; 208: 105735, 2021 04.
Article in English | MEDLINE | ID: mdl-32784045

ABSTRACT

Despite advances in breast cancer (BC) treatment, its mortality remains high due to intrinsic or acquired resistance to therapy. Several ongoing efforts are being made to develop novel drugs to treat this pathology with the aim to overcome resistance, prolong patient survival and improve their quality of life. We have previously shown that the non-hypercalcemic vitamin D analogues EM1 and UVB1 display antitumor effects in preclinical studies employing conventional cell lines and animal models developed from these cells. In this work, we explored the antitumor effects of EM1 and UVB1 employing BC cells derived from patient-derived xenografts (PDXs), which are a powerful preclinical tool for testing new drugs. We demonstrated that the analogues reduced the viability of HER2-positive and Triple Negative BC-PDXs. Moreover, using an in vitro model of acquired resistance to Trastuzumab-emtansine, UVB1 displayed anti-proliferative actions under 2D and 3D culture conditions. It inhibited both formation and growth of established organoids. In addition, a direct correlation between UVB1 antitumor effects and VDR expression in PDXs was found. In conclusion, all the results reinforce the potential use of these vitamin D analogues as antitumor agents to treat HER2-positive and Triple Negative BC.


Subject(s)
Breast Neoplasms/drug therapy , Cell Proliferation/drug effects , Triple Negative Breast Neoplasms/drug therapy , Vitamin D/pharmacology , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Disease Models, Animal , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , Quality of Life , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology , Vitamin D/analogs & derivatives , Xenograft Model Antitumor Assays
4.
J Steroid Biochem Mol Biol ; 185: 118-136, 2019 01.
Article in English | MEDLINE | ID: mdl-30125657

ABSTRACT

Calcitriol analogs have shown promising potential as compounds to be used in cancer chemotherapy. This report presents the synthesis of a novel vitamin D3 derivative with an amide and a carboxyl group in its side chain, called ML-344. In addition, we report its in vitro antitumor activity and its in vivo calcemic effects. We demonstrate that the analog decreases cell viability and retards cell migration of different breast, glioblastoma and head and neck cancer cell lines. Additionally, unlike calcitriol, ML-344 does not display citotoxicity to the murine non-malignant mammary cells and human astrocytes. In concordance with the antimigratory effects found in breast cancer cells, ML-344 decreased the invasive capacity and induced a rearrangement of the actin cytoskeleton in the LM3 breast cancer cell line. In relation to the in vivo studies, the analog did not cause hypercalcemic effects in CF1 mice administered daily at 5 µg/Kg of body weight during a period of 264 h. Finally, computational studies were performed to evaluate the potential binding of the analog to the vitamin D receptor and the in silico assays showed that ML-344 is able to bind to VDR with interesting particularities and greater affinity than calcitriol. Altogether, these results suggest that ML-344 has a promising potential as an antitumor agent with a differential effect between tumor and non-malignant cells.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Calcitriol/analogs & derivatives , Calcitriol/pharmacology , Glioblastoma/drug therapy , Head and Neck Neoplasms/drug therapy , Receptors, Calcitriol/metabolism , Animals , Antineoplastic Agents/chemical synthesis , Astrocytes/drug effects , Calcitriol/chemical synthesis , Cell Line, Tumor , Cell Movement/drug effects , Cell Survival/drug effects , Humans , Male , Mice
5.
Antioxid Redox Signal ; 30(18): 2030-2049, 2019 06 20.
Article in English | MEDLINE | ID: mdl-30484334

ABSTRACT

Aims: Heme oxygenase-1 (HO-1) is an enzyme involved in cellular responses to oxidative stress and has also been shown to regulate processes related to cancer progression. In this regard, HO-1 has been shown to display a dual effect with either antitumor or protumor activity, which is also true for breast cancer (BC). In this work, we address this discrepancy regarding the role of HO-1 in BC. Results: HO-1 was detected in human BC tissues, and its protein levels correlated with reduced tumor size and longer overall survival time of patients, thus suggesting the clinical importance of HO-1 in this type of cancer. Contrariwise, nuclear localization of HO-1 correlated with higher tumor grade suggesting that the effect of HO-1 is dependent on its cellular localization. In vivo experiments showed that both pharmacological activation and genetic overexpression of HO-1 reduce the tumor burden in two different animal models of BC. Furthermore, the pharmacological and genetic activation of HO-1 in several BC cell lines reduce the cellular viability by inducing apoptosis and cell cycle arrest and decrease the cellular migration and invasion rates by modulating pathways involved in the epithelial-mesenchymal transition. Furthermore, HO-1 activation impaired in vivo the metastatic dissemination. Innovation and Conclusion: By using various BC cell lines and animal models as well as human tumor samples, we demonstrated that total HO-1 displays antitumor activities in BC. Furthermore, our study suggests that HO-1 subcellular localization may explain the differential effects observed for the protein in different tumor types.


Subject(s)
Breast Neoplasms/pathology , Cell Nucleus/metabolism , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Up-Regulation , Animals , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Movement , Cell Survival , Epithelial-Mesenchymal Transition , Female , Gene Expression Regulation, Neoplastic , Humans , Mice , Neoplasm Grading , Neoplasm Transplantation , Survival Analysis , Tumor Burden
6.
Oncotarget ; 9(34): 23396-23412, 2018 May 04.
Article in English | MEDLINE | ID: mdl-29805742

ABSTRACT

Triple-negative breast cancer (TNBC) is associated with poor prognosis, high local recurrence rate and high rate of metastasis compared with other breast cancer subtypes. In addition, TNBC lacks a targeted therapy. This scenario highlights the need for novel compounds with high potential for TNBC treatment. In this regard, natural products are important sources of anticancer drugs. D-Fraction, a proteoglucan extracted from the edible and medicinal mushroom Grifola frondosa (Maitake), is a dietary supplement that has been shown to exert both immunostimulatory and immune-independent antitumoral effects on some cancer types. However, its antitumoral potential in TNBC is unknown. Therefore, we employed TNBC cells to investigate if D-Fraction is able to attenuate their aggressive phenotype. We found that D-Fraction decreases MDA-MB-231 cell viability through apoptosis induction and reduces their metastatic potential. D-Fraction increases cell-cell adhesion by increasing E-cadherin protein levels and ß-catenin membrane localization, and increases cell-substrate adhesion. D-Fraction also decreases cell motility by affecting actin cytoskeleton rearrangements, and proteolytic activity of MMP-2 and MMP-9. Furthermore, D-Fraction decreases the invasive capacity of MDA-MB-231 cells. In concordance, D-Fraction retards tumor growth and reduces lung metastases in a xenograft model. Altogether, these results suggest the potential therapeutic role of D-Fraction in aggressive TNBC.

7.
J Steroid Biochem Mol Biol ; 178: 22-35, 2018 04.
Article in English | MEDLINE | ID: mdl-29102624

ABSTRACT

Glioblastoma multiforme (GBM) is the worst and most common brain tumor, characterized by high proliferation and invasion rates. The current standard treatment is mainly based on chemoradiotherapy and this approach has slightly improved patient survival. Thus, novel strategies aimed at prolonging the survival and ensuring a better quality of life are necessary. In the present work, we investigated the antitumoral effect of the novel analogue of calcitriol EM1 on GBM cells employing in vitro, in silico, and in vivo assays. In vitro, we demonstrated that EM1 treatment selectively decreases the viability of murine and human tumor cells without affecting that of normal human astrocytes. The analysis of the mechanisms showed that EM1 produces cell cycle arrest in the T98G cell line, which is accompanied by an increase in p21, p27, p57 protein levels and a decrease in cyclin D1, p-Akt-S473, p-ERK1/2 and c-Jun expression. Moreover, EM1 treatment also exerts in GBM cells anti-migratory effects and decreases their invasive capacity by a reduction in MMP-9 proteolytic activity. In silico, we demonstrated that EM1 is able to bind to the vitamin D receptor with greater affinity than calcitriol. Finally, we showed that EM1 treatment of nude mice administered at 50ug/kg body weight during 21days neither induces hypercalcemia nor toxicity effects. In conclusion, all the results indicate the potential of EM1 analogue as a promising therapeutic alternative for GBM treatment.


Subject(s)
Apoptosis/drug effects , Calcitriol/pharmacology , Cell Cycle/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Glioblastoma/pathology , Animals , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Cell Adhesion/drug effects , Glioblastoma/drug therapy , Humans , Male , Mice , Mice, Nude , Tumor Cells, Cultured , Vitamins/pharmacology
8.
Nutr Cancer ; 69(1): 29-43, 2017 01.
Article in English | MEDLINE | ID: mdl-27892708

ABSTRACT

D-Fraction is protein-bound ß-1,6 and ß-1,3 glucans (proteoglucan) extracted from the edible and medicinal mushroom Grifola frondosa (Maitake). The antitumoral effect of D-Fraction has long been exclusively attributed to their immunostimulatory capacity. However, in recent years increasing evidence showed that D-Fraction directly affects the viability of canine and human tumor cells, independent of the immune system. Previously, we have reported that D-Fraction modulates the expression of genes associated with cell proliferation, cell death, migration, invasion, and metastasis in MCF7 human breast cancer cells. Therefore, the purpose of the current study is to investigate if this modulation of gene expression by Maitake D-Fraction really modulates tumor progression. In the present work, we demonstrate for the first time that Maitake D-Fraction is able to act directly on mammary tumor cells, modulating different cellular processes involved in the development and progression of cancer. We demonstrate that D-Fraction decreases cell viability, increases cell adhesion, and reduces the migration and invasion of mammary tumor cells, generating a less aggressive cell behavior. In concordance with these results, we also demonstrate that D-Fraction decreases tumor burden and the number of lung metastases in a murine model of breast cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Grifola/chemistry , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/pathology , Animals , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Cadherins/metabolism , Cell Adhesion/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Survival/drug effects , Female , Lung Neoplasms/prevention & control , Lung Neoplasms/secondary , Mammary Neoplasms, Experimental/metabolism , Matrix Metalloproteinase 2/metabolism , Mice, Inbred BALB C
9.
J Steroid Biochem Mol Biol ; 163: 193-205, 2016 10.
Article in English | MEDLINE | ID: mdl-27208626

ABSTRACT

Vitamin D has been shown to display a wide variety of antitumour effects, but their therapeutic use is limited by its severe side effects. We have designed and synthesized a Gemini vitamin D analogue of calcitriol (UVB1) which has shown to display antineoplastic effects on different cancer cell lines without causing hypercalcemia. The aim of this work has been to investigate, by employing in silico, in vitro, and in vivo assays, whether UVB1 inhibits human colorectal carcinoma progression. We demonstrated that UVB1 induces apoptotic cell death and retards cellular migration and invasion of HCT116 colorectal carcinoma cells. Moreover, the analogue reduced the tumour volume in vivo, and modulated the expression of Bax, E-cadherin and nuclear ß-catenin in tumour animal tissues without producing toxic effects. In silico analysis showed that UVB1 exhibits greater affinity for the ligand binding domain of vitamin D receptor than calcitriol, and that several characteristics in the three-dimensional conformation of VDR may influence the biological effects. These results demonstrate that the Gemini vitamin D analogue affects the growth of the colorectal cancer and suggest that UVB1 is a potential chemotherapeutic agent for treatment of this disease.


Subject(s)
Antineoplastic Agents/pharmacology , Colorectal Neoplasms/drug therapy , Gene Expression Regulation, Neoplastic , Receptors, Calcitriol/chemistry , Vitamin D/analogs & derivatives , Vitamin D/pharmacology , Animals , Antigens, CD , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Binding Sites , Cadherins/genetics , Cadherins/metabolism , Cell Movement/drug effects , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Disease Progression , HCT116 Cells , Humans , Ligands , Mice , Mice, Nude , Molecular Docking Simulation , Protein Binding , Protein Interaction Domains and Motifs , Receptors, Calcitriol/antagonists & inhibitors , Receptors, Calcitriol/genetics , Receptors, Calcitriol/metabolism , Vitamin D/chemistry , Xenograft Model Antitumor Assays , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism , beta Catenin/genetics , beta Catenin/metabolism
10.
Arch Pharm (Weinheim) ; 348(5): 315-29, 2015 May.
Article in English | MEDLINE | ID: mdl-25864390

ABSTRACT

The active form of vitamin D3, 1α,25(OH)2D3, plays a major role in maintaining calcium/phosphate homeostasis. In addition, it is a potent antiproliferative and pro-differentiating agent. Unfortunately, it usually causes hypercalcemia in vivo when effective antitumour doses are used. It has therefore been found necessary to synthesise new analogues that retain or even increase the antitumour effects but preclude hypercalcemia. This report presents the synthesis of a novel Gemini vitamin D analogue (UVB1) and its biological evaluation. We demonstrate that this compound has potent antitumoural effects over a wide panel of tumour cell lines while showing lack of hypercalcemic activity and toxicity effects in in vivo assays.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Hypercalcemia/chemically induced , Neoplasms/drug therapy , Vitamin D/analogs & derivatives , Vitamin D/pharmacology , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/toxicity , Calcium/blood , Cell Line, Tumor , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Design , Female , Humans , Hypercalcemia/blood , Inhibitory Concentration 50 , Male , Mice , Mice, Nude , Molecular Structure , Neoplasms/pathology , Structure-Activity Relationship , Time Factors , Vitamin D/chemical synthesis , Vitamin D/toxicity
11.
Exp Mol Pathol ; 97(3): 321-31, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25236576

ABSTRACT

The expression of heme oxygenase-1 (HO-1) has been shown to be up-regulated in colorectal cancer (CRC), but the role it plays in this cancer type has not yet been addressed. The aims of this study have been to analyze HO-1 expression in human invasive CRC, evaluate its correlation with clinical and histo-pathological parameters and to investigate the mechanisms through which the enzyme influences tumor progression. We confirmed that HO-1 was over-expressed in human invasive CRC and found that the expression of the enzyme was associated with a longer overall survival time. In addition, we observed in a chemically-induced CRC animal model that total and nuclear HO-1 expression increases with tumor progression. Our investigation of the mechanisms involved in HO-1 action in CRC demonstrates that the protein reduces cell viability through induction of cell cycle arrest and apoptosis and, importantly, that a functional p53 tumor suppressor protein is required for these effects. This reduction in cell viability is accompanied by modulation of the levels of p21, p27, and cyclin D1 and by modulation of Akt and PKC pathways. Altogether, our results demonstrate an antitumoral role of HO-1 and points to the importance of p53 status in this antitumor activity.


Subject(s)
Adenocarcinoma/pathology , Colorectal Neoplasms/pathology , Heme Oxygenase-1/biosynthesis , Tumor Suppressor Protein p53/biosynthesis , Adenocarcinoma/metabolism , Adenocarcinoma/mortality , Animals , Area Under Curve , Blotting, Western , Cell Line, Tumor , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/mortality , Disease Models, Animal , Female , Flow Cytometry , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Male , ROC Curve , Rats , Rats, Wistar , Transfection
12.
Biogerontology ; 8(1): 13-24, 2007 Feb.
Article in English | MEDLINE | ID: mdl-16855859

ABSTRACT

In intestinal cells, 1alpha,25(OH)(2)-vitamin D(3) (1alpha,25(OH)(2)D(3)) regulates gene expression via the specific intracellular vitamin D receptor and induces fast non-transcriptional responses involving stimulation of transmembrane signal transduction pathways. In the present study, we analyzed, for the first time, alterations in p38 MAPK response to 1alpha,25(OH)(2)D(3) in rat enterocytes with ageing. In enterocytes from young rats, the hormone increased, in a time- and dose-dependent fashion, the phosphorylation of p38 MAPK, peaking at 3 min (+2-fold). Basal levels of p38 MAPK phosphorylation were lower in enterocytes from old rats and the hormone response was greatly diminished (+0.5-fold at 3 min). p38 MAPK phosphorylation impairment in old animals was not related to significant changes of the kinase protein expression and do not explain the decreased response to 1alpha,25(OH)(2)D(3). Extracellular and intracellular Ca(2+) chelation or c-Src pharmacological inhibition suppressed hormone activation of p38 MAPK in both, young and aged rats, demonstrating that Ca(2+) and the non-receptor tyrosine kinase c-Src are required for full activation of p38 MAPK in cells stimulated with 1alpha,25(OH)(2)D(3). Two other vitamin D(3) metabolites, 25(OH)D(3) and 24,25(OH)(2)D(3, )also enhanced p38 phosphorylation, and to a similar extent than 1alpha,25(OH)(2)D(3), an ability that is lost with ageing. Enterocyte exposure to the hormone also resulted in the rapid induction of c-fos protein (peaking at 5 min, +3-fold) and to a greater extent than that of mRNA induction. With ageing, 1alpha,25(OH)(2)D(3)-dependent increase of c-fos protein level was diminished, but c-fos mRNA expression was not different from young animals. Impairment of 1alpha,25(OH)(2)D(3) activation of p38 MAPK upon ageing and abnormal hormone regulation of the c-fos oncoprotein synthesis may affect intestinal cell function.


Subject(s)
Aging/metabolism , Calcitriol/pharmacology , Enterocytes/drug effects , Enterocytes/enzymology , p38 Mitogen-Activated Protein Kinases/metabolism , Aging/genetics , Animals , Calcitriol/administration & dosage , Calcium/pharmacology , Dose-Response Relationship, Drug , Duodenum/cytology , Duodenum/drug effects , Duodenum/metabolism , Enterocytes/metabolism , Enzyme Activation/drug effects , Gene Expression/drug effects , Genes, fos , Male , Phosphorylation , Proto-Oncogene Proteins c-fos/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Wistar
13.
J Biol Chem ; 277(38): 35257-62, 2002 Sep 20.
Article in English | MEDLINE | ID: mdl-12124383

ABSTRACT

Sphingosine-1-phosphate (S1P) is a highly bioactive sphingolipid involved in diverse biological processes leading to changes in cell growth, differentiation, motility, and survival. S1P generation is regulated via sphingosine kinase (SK), and many of its effects are mediated through extracelluar action on G-protein-coupled receptors. In this study, we have investigated the mechanisms regulating SK, where this occurs in the cell, and whether this leads to release of S1P extracellularly. The protein kinase C (PKC) activator, phorbol 12-myristate 13-acetate (PMA), induced early activation of SK in HEK 293 cells, and this activation was more specific to the membrane-associated SK. Therefore, we next investigated whether PMA induced translocation of SK to the plasma membrane. PMA induced translocation of both endogenous and green fluorescent protein (GFP)-tagged human SK1 (hSK1) to the plasma membrane. PMA also induced phosphorylation of GFP-hSK1. The PMA-induced translocation was abrogated by preincubation with known PKC inhibitors (bisindoylmaleimide and calphostin-c) as well as by the indirect inhibitor of PKC, C(6)-ceramide, supporting a role for PKC in mediating translocation of SK to the plasma membrane. SK activity was not necessary for translocation, because a dominant negative G82D mutation also translocated in response to PMA. Importantly, PKC regulation of SK was accompanied by a 4-fold increase in S1P in the media. These results demonstrate a novel mechanism by which PKC regulates SK and increases secretion of S1P, allowing for autocrine/paracrine signaling.


Subject(s)
Lysophospholipids , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Protein Kinase C/metabolism , Sphingosine/analogs & derivatives , Sphingosine/metabolism , Tetradecanoylphorbol Acetate/pharmacology , Amino Acid Sequence , Base Sequence , Cell Line , Cell Membrane/enzymology , Cloning, Molecular , DNA Primers , Enzyme Activation , Green Fluorescent Proteins , Humans , Luminescent Proteins/metabolism , Molecular Sequence Data , Phosphotransferases (Alcohol Group Acceptor)/genetics , Protein Transport , Recombinant Fusion Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...