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










Publication year range
1.
Mol Oncol ; 17(7): 1228-1245, 2023 07.
Article in English | MEDLINE | ID: mdl-37081792

ABSTRACT

Cyclin-dependent kinases (CDKs), together with their cyclin partners, are the master cell cycle regulators. Remarkably, the cyclin family was extended to include atypical cyclins, characterized by distinctive structural features, but their partner CDKs remain elusive. Here, we conducted a yeast two-hybrid screen to identify new atypical cyclin-CDK complexes. We identified 10 new complexes, including a complex between CDK6 and cyclin I (CCNI), which was found to be active against retinoblastoma protein. CCNI upregulation increased the proliferation of breast cancer cells in vitro and in vivo, with a magnitude similar to that seen upon cyclin D upregulation, an effect that was abrogated by CDK6 silencing or palbociclib treatment. In line with these findings, CCNI downregulation led to a decrease in cell number and a reduction in the percentage of cells reaching S phase. Finally, CCNI upregulation correlated with the high expression of E2F target genes in large panels of cancer cell lines and tissue samples from breast cancer patients. In conclusion, we unveil CCNI as a new player in the pathways that activate CDK6, enriching the wiring of cell cycle control.


Subject(s)
Breast Neoplasms , Cyclin I , Humans , Female , Cyclin I/genetics , Cyclins/genetics , Cyclins/metabolism , Cell Proliferation/genetics , Breast Neoplasms/genetics , Gene Expression , Cell Cycle Proteins/genetics , Cell Cycle , Cyclin-Dependent Kinase 6/genetics
2.
Cell Rep ; 37(7): 110004, 2021 11 16.
Article in English | MEDLINE | ID: mdl-34788624

ABSTRACT

Polyphosphate (polyP) is a polymer of hundreds of phosphate residues present in all organisms. In mammals, polyP is involved in crucial physiological processes, including coagulation, inflammation, and stress response. However, after decades of research, the metabolic enzymes are still unknown. Here, we purify and identify Nudt3, a NUDIX family member, as the enzyme responsible for polyP phosphatase activity in mammalian cells. We show that Nudt3 shifts its substrate specificity depending on the cation; specifically, Nudt3 is active on polyP when Zn2+ is present. Nudt3 has in vivo polyP phosphatase activity in human cells, and importantly, we show that cells with altered polyP levels by modifying Nudt3 protein amount present reduced viability upon oxidative stress and increased DNA damage, suggesting that polyP and Nudt3 play a role in oxidative stress protection. Finally, we show that Nudt3 is involved in the early stages of embryo development in zebrafish.


Subject(s)
Acid Anhydride Hydrolases/metabolism , Oxidative Stress/physiology , Polyphosphates/metabolism , Acid Anhydride Hydrolases/genetics , Acid Anhydride Hydrolases/physiology , Animals , HEK293 Cells , Humans , Male , Mammals/metabolism , Oxidation-Reduction , Phosphoric Monoester Hydrolases/physiology , Rats , Rats, Sprague-Dawley , Substrate Specificity/physiology , Zebrafish , Zinc/metabolism
3.
Cell Oncol (Dordr) ; 44(6): 1273-1286, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34604945

ABSTRACT

PURPOSE: Cancer stem cells represent a cancer cell subpopulation that has been found to be associated with metastasis and chemoresistance. Therefore, it is vital to identify mechanisms regulating cancer stemness. Previously, we have shown that the atypical cyclin P (CCNP), also known as CNTD2, is upregulated in lung and colorectal cancers and is associated with a worse clinical prognosis. Given that other cyclins have been implicated in pluripotency regulation, we hypothesized that CCNP may also play a role in cancer stemness. METHODS: Cell line-derived spheroids, ex vivo intestinal organoid cultures and induced-pluripotent stem cells (iPSCs) were used to investigate the role of CCNP in stemness. The effects of CCNP on cancer cell stemness and the expression of pluripotency markers and ATP-binding cassette (ABC) transporters were evaluated using Western blotting and RT-qPCR assays. Cell viability was assessed using a MTT assay. The effects of CCNP on WNT targets were monitored by RNA-seq analysis. Data from publicly available web-based resources were also analyzed. RESULTS: We found that CCNP increases spheroid formation in breast, lung and colorectal cancers, and upregulates the expression of stemness (CD44, CD133) and pluripotency (SOX2, OCT4, NANOG) markers. In addition, we found that CCNP promotes resistance to anticancer drugs and induces the expression of multidrug resistance ABC transporters. Our RNA-seq data indicate that CCNP activates the WNT pathway, and that inhibition of this pathway abrogates the increase in spheroid formation promoted by CCNP. Finally, we found that CCNP knockout decreases OCT4 expression in iPSCs, further supporting the notion that CCNP is involved in stemness regulation. CONCLUSION: Our results reveal CCNP as a novel player in stemness and as a potential therapeutic target in cancer.


Subject(s)
Cyclins/metabolism , Neoplastic Stem Cells/metabolism , Wnt Signaling Pathway , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Cyclins/genetics , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Neoplastic Stem Cells/pathology , Pluripotent Stem Cells/metabolism , Wnt Signaling Pathway/genetics
4.
Semin Cell Dev Biol ; 107: 46-53, 2020 11.
Article in English | MEDLINE | ID: mdl-32417219

ABSTRACT

Atypical cyclins have recently emerged as a new subfamily of cyclins characterized by common structural features and interactor pattern. Interestingly, atypical cyclins are phylogenetically close to canonical cyclins, which have well-established roles in cell cycle regulation and cancer. Therefore, although the function of atypical cyclins is still poorly characterized, it seems likely that they are involved in cancer pathogenesis as well. Here, we coupled gene expression and prognostic significance analysis to bibliographic search in order to provide new insights into the role of atypical cyclins in cancer. The information gathered suggests that atypical cyclins intervene in critical processes to sustain cancer growth and have potential to become novel prognostic markers and drug targets in cancer.


Subject(s)
Cyclins/metabolism , Neoplasms/metabolism , Animals , Cell Proliferation/genetics , Cyclins/genetics , Humans , Molecular Targeted Therapy , Neoplasms/genetics , Neoplasms/pathology , Neoplasms/therapy , Prognosis
5.
Cell Mol Life Sci ; 77(2): 231-242, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31420702

ABSTRACT

Regulation of cell division is orchestrated by cyclins, which bind and activate their catalytic workmates, the cyclin-dependent kinases (CDKs). Cyclins have been traditionally defined by an oscillating (cyclic) pattern of expression and by the presence of a characteristic "cyclin box" that determines binding to the CDKs. Noteworthy, the Human Genome Sequence Project unveiled the existence of several other proteins containing the "cyclin box" domain. These potential "cyclins" have been named new, orphan or atypical, creating a conundrum in cyclins nomenclature. Moreover, although many years have passed after their discovery, the scarcity of information regarding these possible members of the family has hampered the establishment of criteria for systematization. Here, we discuss the criteria that define cyclins and we propose a classification and nomenclature update based on structural features, interactors, and phylogenetic information. The application of these criteria allows to systematically define, for the first time, the subfamily of atypical cyclins and enables the use of a common nomenclature for this extended family.


Subject(s)
Cyclins/genetics , Animals , Cell Division/genetics , Cyclin-Dependent Kinases/genetics , Genome, Human/genetics , Humans , Phylogeny
6.
Exp Mol Med ; 51(4): 1-17, 2019 04 16.
Article in English | MEDLINE | ID: mdl-30992425

ABSTRACT

CDK16 (also known as PCTAIRE1 or PCTK1) is an atypical member of the cyclin-dependent kinase (CDK) family that forms an active complex with cyclin Y (CCNY). Although both proteins have been recently implicated in cancer pathogenesis, it is still unclear how the CDK16/CCNY complex exerts its biological activity. To understand the CDK16/CCNY network, we used complementary proteomic approaches to identify potential substrates of this complex. We identified several candidates implicating the CDK16/CCNY complex in cytoskeletal dynamics, and we focused on the microtubule-associated protein regulator of cytokinesis (PRC1), an essential protein for cell division that organizes antiparallel microtubules and whose deregulation may drive genomic instability in cancer. Using analog-sensitive (AS) CDK16 generated by CRISPR-Cas9 mutagenesis in 293T cells, we found that specific inhibition of CDK16 induces PRC1 dephosphorylation at Thr481 and delocalization to the nucleus during interphase. The observation that CDK16 inhibition and PRC1 downregulation exhibit epistatic effects on cell viability confirms that these proteins can act through a single pathway. In conclusion, we identified PRC1 as the first substrate of the CDK16/CCNY complex and demonstrated that the proliferative function of CDK16 is mediated by PRC1 phosphorylation. As CDK16 is emerging as a critical node in cancer, our study reveals novel potential therapeutic targets.


Subject(s)
Cell Cycle Proteins/metabolism , Cyclin-Dependent Kinases/metabolism , Cell Cycle Proteins/genetics , Cell Division/genetics , Cell Division/physiology , Cell Line , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Cyclin-Dependent Kinases/genetics , HEK293 Cells , HeLa Cells , Humans , MCF-7 Cells , Phosphorylation , Protein Binding/genetics , Protein Binding/physiology
7.
Sci Rep ; 8(1): 11797, 2018 08 07.
Article in English | MEDLINE | ID: mdl-30087414

ABSTRACT

Colorectal cancer (CRC) is one of the most common cancers worldwide, with 8-10% of these tumours presenting a BRAF (V600E) mutation. Cyclins are known oncogenes deregulated in many cancers, but the role of the new subfamily of atypical cyclins remains elusive. Here we have performed a systematic analysis of the protein expression levels of eight atypical cyclins in human CRC tumours and several cell lines, and found that CNTD2 is significantly upregulated in CRC tissue compared to the adjacent normal one. CNTD2 overexpression in CRC cell lines increases their proliferation capacity and migration, as well as spheroid formation capacity and anchorage-independent growth. Moreover, CNTD2 increases tumour growth in vivo on xenograft models of CRC with wild-type BRAF. Accordingly, CNTD2 downregulation significantly diminished the proliferation of wild-type BRAF CRC cells, suggesting that CNTD2 may represent a new prognostic factor and a promising drug target in the management of CRC.


Subject(s)
Cell Movement , Cell Proliferation , Colonic Neoplasms/metabolism , Cyclins/metabolism , Mutation, Missense , Proto-Oncogene Proteins B-raf/metabolism , Amino Acid Substitution , Animals , Cell Line, Tumor , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Cyclins/genetics , Female , Humans , Male , Mice , Mice, Nude , Proto-Oncogene Proteins B-raf/genetics
8.
Chem Biol Interact ; 277: 176-184, 2017 Nov 01.
Article in English | MEDLINE | ID: mdl-28947257

ABSTRACT

The activation of the G protein-coupled estrogen receptor (GPER) by its specific agonist G-1 inhibits prostate cancer and 17ß-estradiol-stimulated breast cancer cell proliferation. Tamoxifen (TAM), which also activates the GPER, decreases melanoma cell proliferation, but its action mechanism remains controversial. Here we investigated the expression and the effects of GPER activation by G-1, TAM and its key metabolite endoxifen (EDX) on melanoma cells. Mouse melanoma K1735-M2 cells expressed GPER and G-1 reduced cell biomass, and the number of viable cells, without increasing cell death. Rather, G-1 decreased cell division by blocking cell cycle progression in G2. Likewise, TAM and EDX exhibited an antiproliferative activity in melanoma cells due to decreased cell division. Both G-1 and the antiestrogens showed a trend to decrease the levels of phosphorylated ERK 1/2 after 1 h treatment, although only EDX, the most potent antiproliferative antiestrogen, induced significant effects. Importantly, the targeting of GPER with siRNA abolished the cytostatic activity of both G-1 and antiestrogens, suggesting that the antitumor actions of antiestrogens in melanoma cells involve GPER activation. Our results unveil a new target for melanoma therapy and identify GPER as a key mediator of antiestrogen antiproliferative effects, which may contribute to select the patients that benefit from an antiestrogen-containing regimen.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Cyclopentanes/pharmacology , Melanoma/drug therapy , Quinolines/pharmacology , Receptors, G-Protein-Coupled/agonists , Animals , Cell Line, Tumor , Melanoma/metabolism , Mice , Receptors, Estrogen/analysis , Receptors, Estrogen/metabolism , Receptors, G-Protein-Coupled/analysis , Receptors, G-Protein-Coupled/metabolism
9.
DNA Repair (Amst) ; 57: 171-178, 2017 09.
Article in English | MEDLINE | ID: mdl-28822913

ABSTRACT

Cells require extra amounts of dNTPs to repair DNA after damage. Polyphosphate (polyP) is an evolutionary conserved linear polymer of up to several hundred inorganic phosphate (Pi) residues that is involved in many functions, including Pi storage. In the present article, we report on findings demonstrating that polyP functions as a source of Pi when required to sustain the dNTP increment essential for DNA repair after damage. We show that mutant yeast cells without polyP produce less dNTPs upon DNA damage and that their survival is compromised. In contrast, when polyP levels are ectopically increased, yeast cells become more resistant to DNA damage. More importantly, we show that when polyP is reduced in HEK293 mammalian cell line cells and in human dermal primary fibroblasts (HDFa), these cells become more sensitive to DNA damage, suggesting that the protective role of polyP against DNA damage is evolutionary conserved. In conclusion, we present polyP as a molecule involved in resistance to DNA damage and suggest that polyP may be a putative target for new approaches in cancer treatment or prevention.


Subject(s)
Cell Survival , DNA Damage , DNA Repair , DNA/metabolism , Polyphosphates/metabolism , Deoxyribonucleotides/metabolism , HEK293 Cells , Humans , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae/physiology
10.
Cancer Chemother Pharmacol ; 79(2): 219-225, 2017 02.
Article in English | MEDLINE | ID: mdl-27586965

ABSTRACT

Glutamate has a trophic function in the development of the central nervous system, regulating the proliferation and migration of neuronal progenitors. The resemblance between neuronal embryonic and tumor cells has paved the way for the investigation of the effects of glutamate on tumor cells. Indeed, tumor cells derived from neuronal tissue express ionotropic glutamate receptor (iGluRs) subunits and iGluR antagonists decrease cell proliferation. Likewise, iGluRs subunits are expressed in several peripheral cancer cells and blockade of the N-methyl-D-aspartate (NMDA) and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) ionotropic glutamate receptor subtypes decreases their proliferation and migration. Although these mechanisms are still being investigated, the inhibition of the mitogen-activated protein kinase pathway was shown to play a key role in the antiproliferative activity of iGluR antagonists. Importantly, MK-801, a NMDAR channel blocker, was effective and well tolerated in animal models of melanoma, lung, and breast cancers, suggesting that the blockade of iGluR signaling may represent a new strategy for cancer treatment. In this review, we focus on the significance of NMDA and AMPA receptor expression in tumor cells, as well as possible therapeutic strategies targeting these receptors.


Subject(s)
Neoplasms/drug therapy , Receptors, Ionotropic Glutamate/antagonists & inhibitors , Animals , Cell Movement/drug effects , Cell Proliferation/drug effects , Humans , Receptors, AMPA/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors
11.
Curr Genet ; 63(1): 15-18, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27221322

ABSTRACT

Phosphate is one of the essential elements supporting life. Cells accumulate phosphate in the form of a molecule called polyphosphate (polyP), which carries many functions in the physiology of cells that have not been wholly elucidated. Polyphosphate is present in all the types of cells from bacteria to mammals. It consists of a linear polymer constructed with anywhere from a few to hundreds of inorganic phosphate (Pi) molecules linked by phosphoanhydride bonds. Although polyP was described many years ago, difficulties in the study of its roles, most likely due to the many processes polyP is involved in and incomplete information obtained from multiple models and organisms relegate polyP into oblivion. But now, several interesting pieces of evidence are resurrecting the polyP as a key molecule in processes, such as protein folding, carbon metabolism, cell cycle progression, dNTP synthesis, and genomic stability. In this contribution, in addition to briefly summarize the polyP history and roles, we discuss its involvement in supporting cell cycle progression and genomic stability as well as the implications for the truthful replication of genomes.


Subject(s)
Polyphosphates/metabolism , Bacterial Physiological Phenomena , Cell Cycle , Homeostasis , Polyphosphates/chemistry
12.
Eur J Pharmacol ; 764: 372-378, 2015 Oct 05.
Article in English | MEDLINE | ID: mdl-26165763

ABSTRACT

The use of the antiestrogen tamoxifen in melanoma therapy is controversial due to the unsuccessful outcomes and a still rather unclarified mechanism of action. It seemed that the days of tamoxifen in malignant melanoma therapy were close to an end, but new evidence may challenge this fate. On one hand, it is now believed that metabolism is a major determinant of tamoxifen clinical outcomes in breast cancer patients, which is a variable that has yet to be tested in melanoma patients, since the tamoxifen active metabolite endoxifen demonstrated superior cytostatic activity over the parent drug in melanoma cells; on the other hand, new evidence has emerged regarding estrogen-mediated signaling in melanoma cells, including the methylation of the estrogen receptor-α gene promoter and the expression of the G protein coupled estrogen receptor. The expression of estrogen receptor-α and G protein coupled estrogen receptor, as well as the cytochrome P450 (CYP) 2D6 genotype, may be used as predictive biomarkers to select the patients that may respond to antiestrogens based on specific traits of their tumors. This review focused on these new evidences and how they may contribute to shed new light on this long-lasting controversy, as well as their possible implications for future investigations.


Subject(s)
Antineoplastic Agents, Hormonal/therapeutic use , Estrogen Antagonists/therapeutic use , Melanoma/drug therapy , Skin Neoplasms/drug therapy , Tamoxifen/therapeutic use , Animals , Antineoplastic Agents, Hormonal/adverse effects , Antineoplastic Agents, Hormonal/metabolism , Biotransformation , Cytochrome P-450 CYP2D6/genetics , Cytochrome P-450 CYP2D6/metabolism , Estrogen Antagonists/adverse effects , Estrogen Antagonists/metabolism , Estrogen Receptor alpha/antagonists & inhibitors , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Genotype , Humans , Melanoma/genetics , Melanoma/metabolism , Melanoma/pathology , Receptors, Estrogen/antagonists & inhibitors , Receptors, Estrogen/metabolism , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/drug effects , Skin Neoplasms/genetics , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Tamoxifen/adverse effects , Tamoxifen/analogs & derivatives , Tamoxifen/metabolism
13.
Cancer Lett ; 353(1): 17-24, 2014 Oct 10.
Article in English | MEDLINE | ID: mdl-25042865

ABSTRACT

All-trans-retinoic acid (RA) is a promising agent for breast cancer treatment, but it induces several adverse effects and the few clinical trials performed up to now in breast cancer patients have provided disappointing results. The combination of RA and antiestrogenic compounds, such as tamoxifen, synergistically decreases the proliferation of breast cancer cells and an interplay between retinoid and estrogen signaling has begun to be unraveled, turning these combinations into an appealing strategy for breast cancer treatment. This review focus on the current knowledge regarding the interplay between retinoid and estrogen signaling in breast cancer and the combinations of RA with antiestrogens, aiming their future utilization in cancer therapy.


Subject(s)
Breast Neoplasms/metabolism , Estrogens/metabolism , Retinoids/metabolism , Signal Transduction , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Proliferation , Drug Resistance, Neoplasm , Drug Synergism , Estrogen Receptor Modulators/administration & dosage , Female , Humans , Male , Receptor Cross-Talk , Receptors, Estrogen/metabolism , Receptors, Retinoic Acid/metabolism , Retinoids/administration & dosage , Signal Transduction/drug effects , Tretinoin/administration & dosage
14.
Toxicology ; 323: 10-8, 2014 Sep 02.
Article in English | MEDLINE | ID: mdl-24881593

ABSTRACT

Tamoxifen (TAM) is routinely used in the treatment of breast carcinoma. TAM-induced liver injury remains a major concern, as TAM causes hepatic steatosis in a significant number of patients, which can progress toward steatohepatitis. Liver toxicity is generally believed to involve mitochondrial dysfunction and TAM exerts multiple deleterious effects on mitochondria, which may account for the hepatotoxicity observed in patients treated with TAM. Endoxifen (EDX), a key active metabolite of TAM that is being investigated as an alternative to TAM in breast cancer therapy, slightly affects mitochondria in comparison with TAM and this demonstration well correlates with the absence of alterations in the clinical parameters of individuals taking EDX. The steady-state plasma concentrations of TAM and its active metabolites EDX and 4-hydroxytamoxifen (OHTAM) in patients taking TAM are highly variable, reflecting genetic variants of CYP2D6 involved in TAM metabolism. Besides de genetic polymorphisms, the intake of drugs that influence the enzymatic activity of CYP2D6 compromises the therapeutic efficiency of TAM. The knowledge of the impact of the variability of TAM metabolism in the breast cancer treatment explains the discrepant outcomes observed in patients taking TAM, as well as the individual variability of idiosyncratic liver injury and other sides effects observed. Therefore, and contrarily to the clinical use of EDX, the need of therapeutic drug monitoring and a regular assessment of liver function biomarkers should be considered in patients under therapies with TAM. In this review we focus on the mitochondrial effects of TAM and its metabolites and on the role played by mitochondria in the initiating events leading to TAM-induced hepatotoxicity, as well as the clinical implications.


Subject(s)
Antineoplastic Agents, Hormonal/adverse effects , Chemical and Drug Induced Liver Injury/etiology , Mitochondria, Liver/drug effects , Tamoxifen/adverse effects , Animals , Chemical and Drug Induced Liver Injury/metabolism , Humans , Mitochondria, Liver/metabolism
15.
Exp Cell Res ; 321(2): 288-96, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24240127

ABSTRACT

Recent reports suggest that N-methyl-d-aspartate receptor (NMDAR) blockade by MK-801 decreases tumor growth. Thus, we investigated whether other ionotropic glutamate receptor (iGluR) antagonists were also able to modulate the proliferation of melanoma cells. On the other hand, the antiestrogen tamoxifen (TAM) decreases the proliferation of melanoma cells, and is included in combined therapies for melanoma. As the efficacy of TAM is limited by its metabolism, we investigated the effects of the NMDAR antagonist MK-801 in combination with TAM and its active metabolites, 4-hydroxytamoxifen (OHTAM) and endoxifen (EDX). The NMDAR blockers MK-801 and memantine decreased mouse melanoma K1735-M2 cell proliferation. In contrast, the NMDAR competitive antagonist APV and the AMPA and kainate receptor antagonist NBQX did not affect cell proliferation, suggesting that among the iGluR antagonists only the NMDAR channel blockers inhibit melanoma cell proliferation. The combination of antiestrogens with MK-801 potentiated their individual effects on cell biomass due to diminished cell proliferation, since it decreased the cell number and DNA synthesis without increasing cell death. Importantly, TAM metabolites combined with MK-801 promoted cell cycle arrest in G1. Therefore, the data obtained suggest that the activity of MK-801 and antiestrogens in K1735-M2 cells is greatly enhanced when used in combination.


Subject(s)
Antineoplastic Agents, Hormonal/pharmacology , Cell Proliferation/drug effects , Dizocilpine Maleate/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Melanoma/pathology , Tamoxifen/pharmacology , Animals , Drug Evaluation, Preclinical , Drug Therapy, Combination , Melanoma/drug therapy , Mice , Tamoxifen/analogs & derivatives , Tamoxifen/metabolism , Tumor Cells, Cultured
16.
J Bioenerg Biomembr ; 45(4): 383-96, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23779226

ABSTRACT

The combination of isotretinoin (13-cis-retinoic acid) with antiestrogens seems to be a promising strategy for cancer chemotherapy. The aim of the study was to evaluate the effects of isotretinoin alone or in combination with 4-hydroxytamoxifen (OHTAM) and with its prodrug tamoxifen (TAM), on the functions of rat liver mitochondria, i.e., mitochondrial permeability transition (MPT), bioenergetic functions and adenine nucleotide translocase (ANT). Isotretinoin (5 nmol/mg protein) induced the Ca²âº-dependent MPT pore opening in mitochondria energized with succinate, which was prevented by OHTAM, cyclosporine A, TAM and ANT ligands. When mitochondria were energized with glutamate/malate and in the absence of added Ca²âº isotretinoin decreased the state 3 respiration, the ATP levels, the active ANT content and increased the lag phase of the phosphorylation cycle, demonstrating that isotretinoin decreased the mitochondrial phosphorylation efficiency. These changes of isotretinoin in bioenergetic parameters were not significant in the presence of succinate. The effects of isotretinoin at 5 nmol/mg protein on the Ca²âº-dependent MPT and phosphorylative efficacy may be related with interactions with the ANT. Above 10 nmol/mg protein isotretinoin strongly diminished the active ANT content, decreased the Δψ, inhibited the complex I and induced proton leak through the Fo fraction of complex V. The combination of OHTAM with isotretinoin only induced significant changes in the energy production systems at concentrations ≥5 nmol isotretinoin/mg protein. Therefore, our results suggest that isotretinoin-associated liver toxicity is possibly related with mitochondrial dysfunctions and that the combination with OHTAM may contribute to decrease its toxicity.


Subject(s)
Antineoplastic Agents, Hormonal/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Estrogen Receptor Modulators/pharmacology , Isotretinoin/pharmacology , Mitochondria, Liver/drug effects , Tamoxifen/analogs & derivatives , Tamoxifen/pharmacology , Animals , Antineoplastic Agents, Hormonal/administration & dosage , Cell Membrane Permeability/drug effects , Disease Models, Animal , Drug Interactions , Energy Metabolism , Estrogen Receptor Modulators/administration & dosage , Isotretinoin/administration & dosage , Membrane Potential, Mitochondrial/drug effects , Mitochondria, Liver/metabolism , Mitochondrial ADP, ATP Translocases/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Permeability Transition Pore , Oxidative Phosphorylation , Rats , Rats, Wistar , Tamoxifen/administration & dosage
17.
Life Sci ; 93(2-3): 96-107, 2013 Jul 30.
Article in English | MEDLINE | ID: mdl-23764110

ABSTRACT

AIMS: The clinical utilization of the combinations of all-trans-retinoic acid (RA) with antiestrogens, which present synergism of action in breast cancer, has been limited by RA adverse effects, including hepatotoxicity, which may be related with mitochondrial damage. This work evaluated the effects of RA alone and in combination with the antiestrogen endoxifen (EDX) on liver mitochondria. MAIN METHODS: Mitochondrial permeability transition (MPT) was assessed by using Calcium Green-5N fluorescence and a tetraphenylphosphonium selective electrode. Oxidative stress was evaluated by oxygen consumption and thiobarbituric acid method. Mitochondrial bioenergetic was monitored by measuring oxygen consumption and mitochondrial membrane potential (ΔΨ). Osmotic volume changes of mitochondria were followed at 540nm. KEY FINDINGS: EDX prevents the MPT induced by RA, allowing mitochondria pre-incubated with RA to accumulate Ca(2+) and inhibiting the depolarization of ΔΨ. RA above 10 nmol/mg protein depresses the phosphorylation capacity of mitochondria, as shown by the increase in the time required for ADP phosphorylation as well as by the decrease in state 3 respiration. At 20 nmol/mg protein, RA decreases the ΔΨ and increases the state 4 respiration, suggesting that high concentrations of RA permeabilize the membrane to protons, possibly due to a proton leak through the Fo fraction of complex V. Moreover, the effects of RA on mitochondrial bioenergetics are not changed by EDX. SIGNIFICANCE: RA-induced hepatotoxicity may be related with induction of MPT and alterations in bioenergetic parameters; the combination with EDX, which reduces mitochondrial dysfunction and synergistically potentiates the anticancer activity, may provide a safer therapeutic strategy.


Subject(s)
Mitochondria, Liver/drug effects , Mitochondria, Liver/metabolism , Tamoxifen/analogs & derivatives , Tretinoin/pharmacology , Animals , Calcium/metabolism , Dose-Response Relationship, Drug , Drug Synergism , Energy Metabolism/drug effects , Estrogen Receptor Modulators/pharmacology , Female , Male , Membrane Potential, Mitochondrial/drug effects , Mitochondrial Membrane Transport Proteins/drug effects , Mitochondrial Permeability Transition Pore , Oxidative Stress/drug effects , Phosphorylation/drug effects , Rats , Rats, Wistar , Tamoxifen/pharmacology , Tretinoin/metabolism
18.
Eur J Pharmacol ; 715(1-3): 354-62, 2013 Sep 05.
Article in English | MEDLINE | ID: mdl-23712006

ABSTRACT

Melanoma incidence is dramatically increasing and the available treatments beyond partial efficacy have severe side effects. Retinoids are promising anticancer agents, but their clinical use has been limited by their toxicity, although a combination with other agents can possibly generate a therapeutic action at lower dosage. Thus, we investigated the effects of all-trans-retinoic acid combined with the antiestrogen endoxifen on melanoma cell proliferation and the effects were compared with its pro-drug tamoxifen. Moreover, we evaluated the effects of these combinations on non-neoplasic cells and assessed mitochondrial bioenergetic functions, to predict their potential toxicity. Individually, all-trans-retinoic acid and the antiestrogens endoxifen and tamoxifen decreased melanoma cell biomass, cell viability and DNA synthesis, without increased cell death, suggesting that the compounds inhibited cell proliferation. Noteworthy, endoxifen decreased cell proliferation more efficiently than tamoxifen. The combination of endoxifen with all-trans-retinoic acid enhanced the antiproliferative effects of the compounds individually more potently than tamoxifen, which did not enhance the effects induced by all-trans-retinoic acid alone, and blocked cell cycle progression in G1. Moreover, the combination of all-trans-retinoic acid with endoxifen significantly decreased melanoma cells migration, whereas the combination with tamoxifen did not present significant effects. At the concentrations used the compounds did not induce cytotoxicity in non-neoplasic cells and liver mitochondrial bioenergetic function was not affected. Altogether, our results show for the first time that a combined treatment of all-trans-retinoic acid with endoxifen may provide an anti-proliferative and anti-migration effect upon melanoma cells without major toxicity, offering a powerful therapeutic strategy for malignant melanoma.


Subject(s)
Cell Movement/drug effects , Melanoma/pathology , Tamoxifen/analogs & derivatives , Tretinoin/pharmacology , Tretinoin/toxicity , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Antineoplastic Agents/toxicity , Antineoplastic Combined Chemotherapy Protocols , Cell Line, Tumor , Cell Proliferation/drug effects , Energy Metabolism/drug effects , Estrogen Receptor Modulators/administration & dosage , Estrogen Receptor Modulators/pharmacology , Estrogen Receptor Modulators/toxicity , Female , Liver/cytology , Male , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Rats , Tamoxifen/administration & dosage , Tamoxifen/pharmacology , Tamoxifen/toxicity , Tretinoin/administration & dosage , Xenograft Model Antitumor Assays
19.
Toxicology ; 306: 93-100, 2013 Apr 05.
Article in English | MEDLINE | ID: mdl-23384448

ABSTRACT

Acitretin is a synthetic retinoid used for severe extensive psoriasis and it has been shown to be an effective and a safe therapeutic drug for other diseases including cancer when used in combination with other agents. However, cases of acitretin-associated liver injury have been documented, but the possible mechanisms of acitretin-associated hepatotoxicity and apoptosis are not entirely clarified. This study reports that mitochondrial dysfunctions may play an important role in liver injury and apoptosis induced by this retinoid. Acitretin (5-20 µM) impaired mitochondrial phosphorylation efficiency as demonstrated by the decrease in the state 3 respiration and ATP levels, and by the increase in the lag phase of ADP phosphorylation cycle, without affecting the membrane potential. Acitretin induced Ca(2+)-mediated mitochondrial permeability transition (MPT) and decreased the adenine nucleotide translocase (ANT) content. Acitretin-induced MPT was not prevented by thiol group protecting and antioxidant agents, excluding the involvement of oxidative stress mechanisms. However, MPT was prevented by ANT ligands ATP, ADP, tamoxifen and 4-hydroxytamoxifen, implying that the MPT induction by acitretin is mediated by the ANT. ANT plays a major role in promoting apoptosis and ATP synthesis, and it is still considered as a structural component of the pore with a regulatory role in MPT formation. Therefore, our results, including the decrease in the state 3 respiration and the increase in the lag phase of phosphorylation cycle, the ATP depletion and the induction of Ca(2+)-mediated MPT, indicate that acitretin-associated liver toxicity and apoptosis is possibly related with mitochondrial dysfunctions due to interactions with the ANT. Additionally, the combination of acitretin with other drugs, such as antiestrogens, which are able to inhibit the MPT, may contribute to decrease the toxicity induced by acitretin.


Subject(s)
Acitretin/toxicity , Apoptosis/drug effects , Membrane Potential, Mitochondrial/drug effects , Mitochondria, Liver/drug effects , Mitochondria, Liver/metabolism , Animals , Energy Metabolism/drug effects , Liver/cytology , Liver/drug effects , Liver/metabolism , Mitochondrial ADP, ATP Translocases/metabolism , Phosphorylation/drug effects , Rats , Rats, Wistar , Tamoxifen/analogs & derivatives , Tamoxifen/pharmacology
20.
Toxicol Appl Pharmacol ; 267(1): 104-12, 2013 Feb 15.
Article in English | MEDLINE | ID: mdl-23274567

ABSTRACT

Endoxifen (EDX) is a key active metabolite of tamoxifen (TAM) with higher affinity and specificity to estrogen receptors that also inhibits aromatase activity. It is safe and well tolerated by healthy humans, but its use requires toxicological characterization. In this study, the effects of EDX on mitochondria, the primary targets for xenobiotic-induced toxicity, were monitored to clarify its potential side effects. EDX up to 30 nmol/mg protein did not affect the mitochondrial oxidative phosphorylation. At 50 nmol EDX/mg protein, EDX decreased the ADP phosphorylation rate and a partial collapse of mitochondrial membrane potential (Δψ), that parallels a state 4 stimulation, was observed. As the stimulation of state 4 was not inhibited by oligomycin and 50 nmol EDX/mg protein caused a slight decrease in the light scattering of mitochondria, these data suggest that EDX promotes membrane permeabilization to protons, whereas TAM at the same concentration induced mitochondrial membrane disruption. Moreover, EDX at 10 nmol/mg protein prevented and reversed the Ca(2+)-induced depolarization of ΔΨ and the release of mitochondrial Ca(2+), similarly to cyclosporine A, indicating that EDX did not affect Ca(2+) uptake, but directly interfered with the proteins of the mitochondrial permeability transition (MPT) megacomplex, inhibiting MPT induction. At this concentration, EDX exhibited antioxidant activity that may account for the protective effect against MPT pore opening. In conclusion, EDX within the range of concentrations reached in tissues did not significantly damage the bioenergetic functions of mitochondria, contrarily to the prodrug TAM, and prevented the MPT pore opening and the oxidative stress in mitochondria, supporting that EDX may be a less toxic drug for women with breast carcinoma.


Subject(s)
Estrogen Antagonists/administration & dosage , Intracellular Membranes/drug effects , Mitochondria, Liver/drug effects , Oxidative Stress/drug effects , Porins/metabolism , Tamoxifen/analogs & derivatives , Animals , Female , Intracellular Membranes/metabolism , Male , Mitochondria, Liver/metabolism , Oxidative Stress/physiology , Permeability/drug effects , Phosphorylation/drug effects , Phosphorylation/physiology , Protective Agents/pharmacology , Rats , Rats, Wistar , Tamoxifen/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL
...