Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 22(15)2021 Aug 01.
Article in English | MEDLINE | ID: mdl-34361055

ABSTRACT

Gliomas are solid tumors of the central nervous system (CNS) that originated from different glial cells. The World Health Organization (WHO) classifies these tumors into four groups (I-IV) with increasing malignancy. Glioblastoma (GBM) is the most common and aggressive type of brain tumor classified as grade IV. GBMs are resistant to conventional therapies with poor prognosis after diagnosis even when the Stupp protocol that combines surgery and radiochemotherapy is applied. Nowadays, few novel therapeutic strategies have been used to improve GBM treatment, looking for higher efficiency and lower side effects, but with relatively modest results. The circadian timing system temporally organizes the physiology and behavior of most organisms and daily regulates several cellular processes in organs, tissues, and even in individual cells, including tumor cells. The potentiality of the function of the circadian clock on cancer cells modulation as a new target for novel treatments with a chronobiological basis offers a different challenge that needs to be considered in further detail. The present review will discuss state of the art regarding GBM biology, the role of the circadian clock in tumor progression, and new chrono-chemotherapeutic strategies applied for GBM treatment.


Subject(s)
Brain Neoplasms/drug therapy , Circadian Rhythm/drug effects , Drug Development , Glioblastoma/drug therapy , Pharmaceutical Preparations/administration & dosage , Animals , Humans
2.
FASEB J ; 35(2): e21231, 2021 02.
Article in English | MEDLINE | ID: mdl-33428275

ABSTRACT

Tumors of the nervous system including glioblastoma multiforme (GBM) are the most frequent and aggressive form of brain tumors; however, little is known about the impact of the circadian timing system on the formation, growth, and treatment of these tumors. We investigated day/night differences in tumor growth after injection of A530 glioma cells isolated from malignant peripheral nerve sheath tumor (MPNSTs) of NPcis (Trp53+/- ; Nf1+/- ) mice. Synchronized A530 cell cultures expressing typical glial markers were injected at the beginning of the day or night into the sciatic nerve zone of C57BL/6 mice subject to a 12:12 hours light/dark (LD) cycle or after being released to constant darkness (DD). Tumors generated in animals injected early at night in the LD cycle or in DD showed higher growth rates than in animals injected diurnally. No differences were found when animals were injected at the same time with cultures synchronized 12 hours apart. Similar experiments performed with B16 melanoma cells showed higher tumor growth rates in animals injected at the beginning of the night compared to those injected in the daytime. A higher tumor growth rate than that in controls was observed when mice were injected with knocked-down clock gene Bmal1 cells. Finally, when we compared day/night administration of different doses of the proteasome inhibitor Bortezomib (0.5-1.5 mg/kg) in tumor-bearing animals, we found that low-dose chemotherapy displayed higher efficacy when administered at night. Results suggest the existence of a precise temporal control of tumor growth and of drug efficacy in which the host state and susceptibility are critical.


Subject(s)
Brain Neoplasms/pathology , Circadian Rhythm , Glioblastoma/pathology , Photoperiod , Xenograft Model Antitumor Assays/methods , ARNTL Transcription Factors/genetics , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/therapeutic use , Bortezomib/administration & dosage , Bortezomib/therapeutic use , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Cell Line, Tumor , Drug Administration Schedule , Glioblastoma/drug therapy , Glioblastoma/genetics , Mice , Mice, Inbred C57BL , Neurofibromin 1/genetics , Tumor Cells, Cultured , Tumor Suppressor Protein p53/genetics , Xenograft Model Antitumor Assays/standards
3.
Photodiagnosis Photodyn Ther ; 33: 102097, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33232818

ABSTRACT

Glioblastoma is the most severe form of brain cancer. Despite multimodal therapy combining surgery, radiotherapy and chemotherapy, prognosis of patients is dismal. It has been observed that the surgical resection guided by photosensitizer fluorescence followed by photodynamic therapy (PDT) prolongs the average survival in patients with glioblastoma. The main problem with all oncological treatments, including PDT, is the presence of resistant cells. The objective of this study was to isolate and perform an initial characterization of human glioblastoma cells resistant to PDT employing methyl-5-aminolevulinic acid. We obtained resistant cells from the T98 G cell line. Resistant populations accumulated less photosensitizer, formed spheroids of higher number of cells, had higher tumorigenic capacity, and expressed higher mRNA levels of fibroblastic growth factor receptor (FGFR), epidermal growth factor receptor (EGFR) and ß-platelet-derived growth factor receptor (ßPDGFR) than parental cells. The studies of glioblastoma resistance to PDT would help to better understand the causes of tumor recurrence after PDT and to develop new therapeutic proposals in this field of oncology.


Subject(s)
Glioblastoma , Photochemotherapy , Aminolevulinic Acid/pharmacology , Aminolevulinic Acid/therapeutic use , Cell Line, Tumor , Glioblastoma/drug therapy , Humans , Neoplasm Recurrence, Local , Photochemotherapy/methods , Photosensitizing Agents/pharmacology , Photosensitizing Agents/therapeutic use
4.
Biochem J ; 477(23): 4675-4688, 2020 12 11.
Article in English | MEDLINE | ID: mdl-33211090

ABSTRACT

Glioblastoma multiforme is the most aggressive type of tumor of the CNS with an overall survival rate of approximately one year. Since this rate has not changed significantly over the last 20 years, the development of new therapeutic strategies for the treatment of these tumors is peremptory. The over-expression of the proto-oncogene c-Fos has been observed in several CNS tumors including glioblastoma multiforme and is usually associated with a poor prognosis. Besides its genomic activity as an AP-1 transcription factor, this protein can also activate phospholipid synthesis by a direct interaction with key enzymes of their metabolic pathways. Given that the amino-terminal portion of c-Fos (c-Fos-NA: amino acids 1-138) associates to but does not activate phospholipid synthesizing enzymes, we evaluated if c-Fos-NA or some shorter derivatives are capable of acting as dominant-negative peptides of the activating capacity of c-Fos. The over-expression or the exogenous administration of c-Fos-NA to cultured T98G cells hampers the interaction between c-Fos and PI4K2A, an enzyme activated by c-Fos. Moreover, it was observed a decrease in tumor cell proliferation rates in vitro and a reduction in tumor growth in vivo when a U87-MG-generated xenograft on nude mice is intratumorally treated with recombinant c-Fos-NA. Importantly, a smaller peptide of 92 amino acids derived from c-Fos-NA retains the capacity to interfere with tumor proliferation in vitro and in vivo. Taken together, these results support the use of the N-terminal portion of c-Fos, or shorter derivatives as a novel therapeutic strategy for the treatment of glioblastoma multiforme.


Subject(s)
Cell Proliferation , Glioblastoma/metabolism , Minor Histocompatibility Antigens/metabolism , Phospholipids/biosynthesis , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Cell Line, Tumor , Enzyme Activation , Glioblastoma/genetics , Glioblastoma/pathology , Humans , Minor Histocompatibility Antigens/genetics , Phospholipids/genetics , Phosphotransferases (Alcohol Group Acceptor)/genetics , Proto-Oncogene Mas , Proto-Oncogene Proteins c-fos/genetics , Transcription Factor AP-1/genetics , Transcription Factor AP-1/metabolism
5.
Front Cell Neurosci ; 13: 198, 2019.
Article in English | MEDLINE | ID: mdl-31133814

ABSTRACT

The mechanisms that coordinately regulate lipid synthesis in the nervous system together with the high rates of membrane biogenesis needed to support cell growth are largely unknown as are their subcellular site of synthesis. c-Fos, a well-known AP-1 transcription factor, has emerged as a unique protein with the capacity to associate to specific enzymes of the pathway of synthesis of phospholipids at the endoplasmic reticulum and activate their synthesis to accompany genomic decisions of growth. Herein, we discuss this effect of c-Fos in the context of neuronal differentiation and also with respect to pathologies of the nervous system such as the development and growth of tumors. We also provide insights into the sub-cellular sites where this regulation occurs at the endoplasmic reticulum membranes and the molecular mechanism by which c-Fos exerts this activity.

6.
Clin Cancer Res ; 25(13): 4049-4062, 2019 07 01.
Article in English | MEDLINE | ID: mdl-30890549

ABSTRACT

PURPOSE: BRCA1 and BRCA2 deficiencies are widespread drivers of human cancers that await the development of targeted therapies. We aimed to identify novel synthetic lethal relationships with therapeutic potential using BRCA-deficient isogenic backgrounds. EXPERIMENTAL DESIGN: We developed a phenotypic screening technology to simultaneously search for synthetic lethal (SL) interactions in BRCA1- and BRCA2-deficient contexts. For validation, we developed chimeric spheroids and a dual-tumor xenograft model that allowed the confirmation of SL induction with the concomitant evaluation of undesired cytotoxicity on BRCA-proficient cells. To extend our results using clinical data, we performed retrospective analysis on The Cancer Genome Atlas (TCGA) breast cancer database. RESULTS: The screening of a kinase inhibitors library revealed that Polo-like kinase 1 (PLK1) inhibition triggers strong SL induction in BRCA1-deficient cells. Mechanistically, we found no connection between the SL induced by PLK1 inhibition and PARP inhibitors. Instead, we uncovered that BRCA1 downregulation and PLK1 inhibition lead to aberrant mitotic phenotypes with altered centrosomal duplication and cytokinesis, which severely reduced the clonogenic potential of these cells. The penetrance of PLK1/BRCA1 SL interaction was validated using several isogenic and nonisogenic cellular models, chimeric spheroids, and mice xenografts. Moreover, bioinformatic analysis revealed high-PLK1 expression in BRCA1-deficient tumors, a phenotype that was consistently recapitulated by inducing BRCA1 deficiency in multiple cell lines as well as in BRCA1-mutant cells. CONCLUSIONS: We uncovered an unforeseen addiction of BRCA1-deficient cancer cells to PLK1 expression, which provides a new means to exploit the therapeutic potential of PLK1 inhibitors in clinical trials, by generating stratification schemes that consider this molecular trait in patient cohorts.


Subject(s)
BRCA1 Protein/deficiency , Cell Cycle Proteins/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , Synthetic Lethal Mutations/drug effects , Animals , Apoptosis/drug effects , Apoptosis/genetics , BRCA2 Protein/deficiency , BRCA2 Protein/genetics , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Line, Tumor , Cells, Cultured , Chromosome Aberrations , DNA Damage , Disease Models, Animal , Gene Expression , Gene Knockdown Techniques , Humans , Mice , Xenograft Model Antitumor Assays , Polo-Like Kinase 1
7.
Sci Rep ; 9(1): 3010, 2019 02 28.
Article in English | MEDLINE | ID: mdl-30816179

ABSTRACT

Glioblastoma multiforme is considered to be one of the most aggressive types of tumors of the central nervous system, with a poor prognosis and short survival periods of ~ one year. The current protocol for glioblastoma treatment includes the surgical excision of the primary tumor followed by radio and chemotherapy. Photodynamic therapy (PDT) is considered a promising strategy for the treatment of several types of tumors. Phthalocyanines (Pcs) are good photosensitizers (PSs) for PDT because they induce cell death in several cellular models. ZnPc (Zn(II)phthalocyanine) is a well-known Pc, extensively tested in different cells and tumor models, but its evaluation on a glioblastoma model has been poorly studied. Herein, we compare the capacity of ZnPc and one of its derivatives, Zn(II)tetraminephthalocyanine (TAZnPc), to photoinactivate glioblastoma cells (T98G, MO59, LN229 and U87-MG) in culture. We measured the cellular uptake, the toxicity in the dark and the subcellular localization of the different Pcs, as well as the clonogenic capacity of surviving cells after PDT. The mechanism of cell death induced after PDT was determined by measuring caspase 3 activation, DNA fragmentation, phosphatidylserine externalization, mitochondrial morphological changes and loss of mitochondrial membrane potential as well as lysosomal membrane integrity. Overall, ZnPc and TAZnPc present good properties to be used as PSs with photoinactivation capacity on glioblastoma cells.


Subject(s)
Glioblastoma/drug therapy , Indoles/toxicity , Organometallic Compounds/toxicity , Photochemotherapy/methods , Photosensitizing Agents/toxicity , Caspase 3/metabolism , Cell Line, Tumor , DNA Fragmentation , Humans , Indoles/chemistry , Indoles/pharmacology , Isoindoles , Lysosomes/drug effects , Mitochondria/drug effects , Organometallic Compounds/chemistry , Organometallic Compounds/pharmacology , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacology , Zinc Compounds
9.
BMC Cancer ; 15: 761, 2015 Oct 22.
Article in English | MEDLINE | ID: mdl-26493452

ABSTRACT

BACKGROUND: Microcalcifications can be the early and only presenting sign of breast cancer. One shared characteristic of breast cancer is the appearance of mammographic mammary microcalcifications that can routinely be used to detect breast cancer in its initial stages, which is of key importance due to the possibility that early detection allows the application of more conservative therapies for a better patient outcome. The mechanism by which mammary microcalcifications are formed is still largely unknown but breast cancers presenting microcalcifications are more often associated with a poorer prognosis. METHODS: We combined Capillary Electrochromatography, histology, and gene expression (qRT-PCR) to analyze patient-matched normal breast tissue vs. breast tumor. Potential carcinogenicity of oxalate was tested by its inoculation into mice. All data were subjected to statistical analysis. RESULTS: To study the biological significance of oxalates within the breast tumor microenvironment, we measured oxalate concentration in both human breast tumor tissues and adjoining non-pathological breast tissues. We found that all tested breast tumor tissues contain a higher concentration of oxalates than their counterpart non-pathological breast tissue. Moreover, it was established that oxalate induces proliferation of breast cells and stimulates the expression of a pro-tumorigenic gene c-fos. Furthermore, oxalate generates highly malignant and undifferentiated tumors when it was injected into the mammary fatpad in female mice, but not when injected into their back, indicating that oxalate does not induce cancer formation in all types of tissues. Moreover, neither human kidney-epithelial cells nor mouse fibroblast cells proliferate when are treated with oxalate. CONCLUSIONS: We found that the chronic exposure of breast epithelial cells to oxalate promotes the transformation of breast cells from normal to tumor cells, inducing the expression of a proto-oncogen as c-fos and proliferation in breast cancer cells. Furthermore, oxalate has a carcinogenic effect when injected into the mammary fatpad in mice, generating highly malignant and undifferentiated tumors with the characteristics of fibrosarcomas of the breast. As oxalates seem to promote these differences, it is expected that a significant reduction in the incidence of breast cancer tumors could be reached if it were possible to control oxalate production or its carcinogenic activity.


Subject(s)
Breast Neoplasms/etiology , Calcinosis/complications , Mammary Glands, Human/pathology , Mammary Neoplasms, Animal/pathology , Neoplasms, Experimental , Oxalates/metabolism , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Calcinosis/metabolism , Calcinosis/pathology , Cell Line, Tumor , Cell Transformation, Neoplastic , Female , Humans , Mammary Glands, Human/metabolism , Mammary Neoplasms, Animal/metabolism , Mice , Mice, Inbred BALB C , Prognosis
10.
J Biol Chem ; 290(49): 29578-92, 2015 Dec 04.
Article in English | MEDLINE | ID: mdl-26475860

ABSTRACT

Phosphatidic acid (PA) is a central precursor for membrane phospholipid biosynthesis. The lipin family is a magnesium-dependent type I PA phosphatase involved in de novo synthesis of neutral lipids and phospholipids. The regulation of lipin activity may govern the pathways by which these lipids are synthesized and control the cellular levels of important signaling lipids. Moreover, the proto-oncoprotein c-Fos has an emerging role in glycerolipid synthesis regulation; by interacting with key synthesizing enzymes it is able to increase overall phospho- and glycolipid synthesis. We studied the lipin 1ß enzyme activity in a cell-free system using PA/Triton X-100 mixed micelles as substrate, analyzing it in the presence/absence of c-Fos. We found that lipin 1ß kcat value increases around 40% in the presence of c-Fos, with no change in the lipin 1ß affinity for the PA/Triton X-100 mixed micelles. We also probed a physical interaction between both proteins. Although the c-Fos domain involved in lipin activation is its basic domain, the interaction domain is mapped to the N-terminal c-Fos. In conclusion, we provide evidence for a novel positive regulator of lipin 1ß PA phosphatase activity that is not achieved via altering its subcellular localization or affinity for membranes but rather through directly increasing its catalytic efficiency.


Subject(s)
Nuclear Proteins/metabolism , Phosphatidate Phosphatase/metabolism , Proto-Oncogene Proteins c-fos/metabolism , 3T3 Cells , Animals , Cell Nucleus/metabolism , Cytoplasm/metabolism , Diglycerides/chemistry , Fluorescence Resonance Energy Transfer , Gene Deletion , Humans , Lipids/chemistry , Mice , Micelles , Organic Chemicals/chemistry , Phosphatidic Acids/chemistry , Phospholipids/chemistry , Point Mutation , Protein Binding , Protein Structure, Tertiary , Recombinant Proteins/metabolism
11.
Oncotarget ; 6(19): 16883-901, 2015 Jul 10.
Article in English | MEDLINE | ID: mdl-26143639

ABSTRACT

c-Fos is a proto-oncogene involved in diverse cellular functions. Its deregulation has been associated to abnormal development and oncogenic progression. c-fos-/- mice are viable but present a reduction in their body weight and brain size. We examined the importance of c-Fos during neocortex development at 13.5, 14.5 and 16.5 days of gestation. At E14.5, neocortex thickness, apoptosis, mitosis and expression of markers along the different stages of Neural Stem Progenitor Cells (NSPCs) differentiation in c-fos-/- and wild-type mice were analyzed. A ~15% reduction in the neocortex thickness of c-fos-/- embryos was observed which correlates with a decrease in the number of differentiated cells and an increase in apoptosis at the ventricular zone. No difference in mitosis rate was observed, although the mitotic angle was predominantly vertical in c-fos-/- embryos, suggesting a reduced trend of NSPCs to differentiate. At E13.5, changes in differentiation markers start to be apparent and are still clearly observed at E16.5. A tendency of more AP-1/DNA complexes present in nuclear extracts of cerebral cortex from c-fos-/- embryos with no differences in the lipid synthesis activity was found. These results suggest that c-Fos is involved in the normal development of NSPCs by means of its AP-1 activity.


Subject(s)
Cell Differentiation/genetics , Genes, fos/genetics , Neocortex/embryology , Neural Stem Cells/cytology , Neurogenesis/genetics , Animals , Electrophoretic Mobility Shift Assay , Embryo, Mammalian , Fluorescent Antibody Technique , Mice , Mice, Inbred C57BL , Mice, Knockout
12.
Biochim Biophys Acta ; 1841(9): 1241-6, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24886961

ABSTRACT

The mechanisms that co-ordinately activate lipid synthesis when high rates of membrane biogenesis are needed to support cell growth are largely unknown. c-Fos, a well known AP-1 transcription factor, has emerged as a unique protein with the capacity to associate to specific enzymes of the pathway of synthesis of phospholipids at the endoplasmic reticulum and activate their synthesis to accompany genomic decisions of growth. Herein, we discuss this cytoplasmic, non-genomic effect of c-Fos in the context of other mechanisms that have been proposed to regulate lipid synthesis.


Subject(s)
Endoplasmic Reticulum/metabolism , Eukaryotic Cells/metabolism , Phospholipids/biosynthesis , Phosphotransferases (Alcohol Group Acceptor)/genetics , Proto-Oncogene Proteins c-fos/genetics , Animals , Cell Cycle , Cell Proliferation , Eukaryotic Cells/cytology , Gene Expression Regulation , Humans , Lipid Metabolism , Minor Histocompatibility Antigens , Phospholipids/genetics , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Protein Binding , Proto-Oncogene Proteins c-fos/metabolism , Signal Transduction
13.
Biochem J ; 461(3): 521-30, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24819416

ABSTRACT

c-Fos is a well-recognized member of the AP-1 (activator protein-1) family of transcription factors. In addition to this canonical activity, we previously showed that cytoplasmic c-Fos activates phospholipid synthesis through a mechanism independent of its genomic AP-1 activity. c-Fos associates with particular enzymes of the lipid synthesis pathway at the endoplasmic reticulum and increases the Vmax of the reactions without modifying the Km values. This lipid synthesis activation is associated with events of differentiation and proliferation that require high rates of membrane biogenesis. Since lipid synthesis also occurs in the nucleus, and different phospholipids have been assigned transcription regulatory functions, in the present study we examine if c-Fos also acts as a regulator of phospholipid synthesis in the nucleus. Furthermore, we examine if c-Fos modulates transcription through its phospholipid synthesis activator capacity. We show that nuclear-localized c-Fos associates with and activates PI4P5K (phosphatidylinositol-4-monophosphate 5-kinase), but not with PI4KIIIß (type IIIß phosphatidylinositol 4-kinase) thus promoting PtdIns(4,5)P2 (phosphatidylinositol 4,5-bisphosphate) formation, which, in turn, promotes transcriptional changes. We propose c-Fos as a key regulator of nuclear PtdIns(4,5)P2 synthesis in response to growth signals that results in c-Fos-dependent transcriptional changes promoted by the newly synthesized lipids.


Subject(s)
Cell Nucleus/metabolism , Phosphatidylinositol 4,5-Diphosphate/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Transcription, Genetic , Up-Regulation , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cell Nucleus/drug effects , Cell Nucleus/enzymology , Cell Nucleus/ultrastructure , Cell Nucleus Size/drug effects , Cell Proliferation/drug effects , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mice , NIH 3T3 Cells , Nuclear Matrix-Associated Proteins/genetics , Nuclear Matrix-Associated Proteins/metabolism , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Phosphotransferases (Alcohol Group Acceptor)/chemistry , Phosphotransferases (Alcohol Group Acceptor)/genetics , Protein Transport/drug effects , Proto-Oncogene Proteins c-fos/antagonists & inhibitors , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-jun/genetics , Proto-Oncogene Proteins c-jun/metabolism , RNA Interference , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Transcription, Genetic/drug effects , Up-Regulation/drug effects
14.
Phytochemistry ; 94: 260-7, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23827326

ABSTRACT

Sixteen dihydro-ß-agarofuran sesquiterpenes were isolated from the aerial parts of Schaefferia argentinensis Speg. Their structures were determined by a combination of 1D and 2D NMR and MS techniques. The in vitro antiproliferative activity of the major sesquiterpenes was examined in T47D, MCF7, and MDA-MB231 human cancer cell lines, but was found to be marginal.


Subject(s)
Celastraceae/chemistry , Plant Components, Aerial/chemistry , Sesquiterpenes/chemistry , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/isolation & purification , Antineoplastic Agents, Phytogenic/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Humans , MCF-7 Cells , Magnetic Resonance Spectroscopy , Mass Spectrometry/methods , Molecular Structure , Sesquiterpenes/isolation & purification , Sesquiterpenes/pharmacology
15.
IUBMB Life ; 65(7): 584-92, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23712998

ABSTRACT

Lipid synthesis is a complex process regulated at multiple levels. Here, we will discuss nongenomic regulatory mechanisms, particularly the activation and/or recruitment of key enzymes to membranes. The phospholipid synthesis enzymes Lipin and CTP:phosphocholine cytidylyltransferase are taken as examples of these mechanisms that are mediated by posttranslational modifications or by an intrinsic property of the enzyme that senses lipid composition. In addition, special emphasis will be put on another relevant non genomic lipid synthesis regulation mechanism that is dependent on c-Fos, a protein that has deserved less attention so far. This latter regulatory mechanism is emerging as an important determinant for processes that require high rates of lipid synthesis such as those of growth and proliferation.


Subject(s)
Choline-Phosphate Cytidylyltransferase/metabolism , Phospholipids/biosynthesis , Protein Processing, Post-Translational , Cell Membrane/enzymology , Cell Proliferation , Humans , Lipids/biosynthesis , Organic Chemicals/metabolism , Proto-Oncogene Proteins c-fos/biosynthesis , Proto-Oncogene Proteins c-fos/metabolism
16.
PLoS One ; 8(1): e53211, 2013.
Article in English | MEDLINE | ID: mdl-23301044

ABSTRACT

A shared characteristic of tumor cells is their exacerbated growth. Consequently, tumor cells demand high rates of phospholipid synthesis required for membrane biogenesis to support their growth. c-Fos, in addition to its AP-1 transcription factor activity, is the only protein known up to date that is capable of activating lipid synthesis in normal and brain tumor tissue. For this latter activity, c-Fos associates to the endoplasmic reticulum (ER) through its N-terminal domain and activates phospholipid synthesis, an event that requires it Basic Domain (BD) (aa 139-159). Fra-1, another member of the FOS family of proteins, is over-expressed in human breast cancer cells and its BD is highly homologous to that of c-Fos with two conservative substitutions in its basic amino acids. Consequently, herein we examined if Fra-1 and/or c-Fos participate in growth of breast cancer cells by activating phospholipid synthesis as found previously for c-Fos in brain tumors. We found both Fra-1 and c-Fos over-expressed in >95% of human ductal breast carcinoma biopsies examined contrasting with the very low or undetectable levels in normal tissue. Furthermore, both proteins associate to the ER and activate phospholipid synthesis in cultured MCF7 and MDA-MB231 breast cancer cells and in human breast cancer samples. Stripping tumor membranes of Fra-1 and c-Fos prior to assaying their lipid synthesis capacity in vitro results in non-activated lipid synthesis levels that are restored to their initial activated state by addition of Fra-1 and/or c-Fos to the assays. In MDA-MB231 cells primed to proliferate, blocking Fra-1 and c-Fos with neutralizing antibodies blocks lipid-synthesis activation and cells do not proliferate. Taken together, these results disclose the cytoplasmic activity of Fra-1 and c-Fos as potential targets for controlling growth of breast carcinomas by decreasing the rate of membrane biogenesis required for growth.


Subject(s)
Breast Neoplasms/metabolism , Cell Membrane/metabolism , Cytoplasm/metabolism , Gene Expression Regulation, Neoplastic , Proto-Oncogene Proteins c-fos/physiology , Adult , Aged , Aged, 80 and over , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation , Female , Humans , Middle Aged , Phospholipids/chemistry , Protein Structure, Tertiary , Proto-Oncogene Proteins c-fos/metabolism
17.
Neurochem Res ; 37(6): 1364-71, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22476983

ABSTRACT

Some 20 years ago c-Fos was identified as a member of the AP-1 family of inducible transcription factors (Angel and Karin in Biochim Biophys Acta 1072:129-157, 1991). More recently, an additional activity was described for this protein: it associates to the endoplasmic reticulum and activates the biosynthesis of phospholipids (Bussolino et al. in FASEB J 15:556-558, 2001), (Gil et al. in Mol Biol Cell 15:1881-1894, 2004), the quantitatively most important components of cellular membranes. This latter activity of c-Fos determines the rate of membrane genesis and consequently of growth in differentiating PC12 cells (Gil et al. in Mol Biol Cell 15:1881-1894, 2004). In addition, it has been shown that c-Fos is over-expressed both in PNS and CNS tumors (Silvestre et al. in PLoS One 5(3):e9544, 2010). Herein, it is shown that c-Fos-activated phospholipid synthesis is required to support membrane genesis during the exacerbated growth characteristic of brain tumor cells. Specifically blocking c-Fos-activated phospholipid synthesis significantly reduces proliferation of tumor cells in culture. Blocking c-Fos expression also prevents tumor progression in mice intra-cranially xeno-grafted human brain tumor cells. In NPcis mice, an animal model of the human disease Neurofibromatosis Type I (Cichowski and Jacks in Cell 104:593-604, 2001), animals spontaneously develop tumors of the PNS and the CNS, provided they express c-Fos (Silvestre et al. in PLoS One 5(3):e9544, 2010). Treatment of PNS tumors with an antisense oligonucleotide that specifically blocks c-Fos expression also blocks tumor growth in vivo. These results disclose cytoplasmic c-Fos as a new target for effectively controlling brain tumor growth.


Subject(s)
Cell Proliferation/drug effects , Central Nervous System Neoplasms/pathology , Peripheral Nervous System Neoplasms/pathology , Phospholipids/biosynthesis , Proto-Oncogene Proteins c-fos/metabolism , Animals , Cell Line, Tumor , Central Nervous System/metabolism , Central Nervous System Neoplasms/metabolism , Endoplasmic Reticulum/metabolism , Humans , Mice , Oligonucleotides, Antisense/metabolism , PC12 Cells , Peripheral Nervous System Neoplasms/metabolism , Rats
18.
Mol Biol Cell ; 22(24): 4716-25, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21998197

ABSTRACT

The oncoprotein c-Fos is a well-recognized AP-1 transcription factor. In addition, this protein associates with the endoplasmic reticulum and activates the synthesis of phospholipids. However, the mechanism by which c-Fos stimulates the synthesis of phospholipids in general and the specific lipid pathways activated are unknown. Here we show that induction of quiescent cells to reenter growth promotes an increase in the labeling of polyphosphoinositides that depends on the expression of c-Fos. We also investigated whether stimulation by c-Fos of the synthesis of phosphatidylinositol and its phosphorylated derivatives depends on the activation of enzymes of the phosphatidylinositolphosphate biosynthetic pathway. We found that c-Fos activates CDP-diacylglycerol synthase and phosphatidylinositol (PtdIns) 4-kinase II α in vitro, whereas no activation of phosphatidylinositol synthase or of PtdIns 4-kinase II ß was observed. Both coimmunoprecipitation and fluorescence resonance energy transfer experiments consistently showed a physical interaction between the N-terminal domain of c-Fos and the enzymes it activates.


Subject(s)
Phosphatidylinositol Phosphates/biosynthesis , Proto-Oncogene Proteins c-fos/metabolism , Transcription Factor AP-1/metabolism , 1-Phosphatidylinositol 4-Kinase/biosynthesis , 1-Phosphatidylinositol 4-Kinase/genetics , Animals , Diacylglycerol Cholinephosphotransferase/biosynthesis , Diacylglycerol Cholinephosphotransferase/genetics , Enzyme Activation/physiology , Enzyme Induction/physiology , Mice , NIH 3T3 Cells , Phosphatidylinositol Phosphates/genetics , Phospholipids/biosynthesis , Phospholipids/genetics , Protein Structure, Tertiary/physiology , Proto-Oncogene Proteins c-fos/genetics , Transcription Factor AP-1/genetics
19.
PLoS One ; 5(3): e9544, 2010 Mar 04.
Article in English | MEDLINE | ID: mdl-20209053

ABSTRACT

BACKGROUND: We have previously shown that the transcription factor c-Fos is also capable of associating to endoplasmic reticulum membranes (ER) and activating phospholipid synthesis. Herein we examined phospholipid synthesis status in brain tumors from human patients and from NPcis mice, an animal model of the human disease Neurofibromatosis Type 1 (NF1). PRINCIPAL FINDINGS: In human samples, c-Fos expression was at the limit of detection in non-pathological specimens, but was abundantly expressed associated to ER membranes in tumor cells. This was also observed in CNS of adult tumor-bearing NPcis mice but not in NPcis fos(-/-) KO mice. A glioblastoma multiforme and a malignant PNS tumor from a NF1 patient (MPNST) showed a 2- and 4- fold c-Fos-dependent phospholipid synthesis activation, respectively. MPNST samples also showed increased cell proliferation rates and abundant c-Fos expression. CONCLUSIONS: Results highlight a role of cytoplasmic c-Fos as an activator of phospholipid synthesis in events demanding high rates of membrane biogenesis as occurs for the exacerbated growth of tumors cells. They also disclose this protein as a potential target for controlling tumor growth in the nervous system.


Subject(s)
Central Nervous System Neoplasms/pathology , Cytoplasm/metabolism , Proto-Oncogene Proteins c-fos/biosynthesis , Animals , Brain/metabolism , Brain/pathology , Cell Proliferation , Central Nervous System Neoplasms/metabolism , Disease Models, Animal , Endoplasmic Reticulum/metabolism , Genotype , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurofibromatosis 1/metabolism , Phosphorylation
20.
Biopolymers ; 91(9): 710-8, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19384981

ABSTRACT

This work explores the surface properties of the transcription factor Fra-1 and compares them with those of two other immediate early proteins, c-Fos and c-Jun, to establish generalities and differences in the surface behavior and interaction with phospholipids of this type of proteins. We present several experimental clues of the flexible nature of Fra-1, c-Fos, and c-Jun that support sequence-based predictions of their intrinsical disorder. The values of surface parameters for Fra-1 are similar in general to those of c-Fos and c-Jun. However, we find differences in the interactions of the three proteins with phospholipids. The closely related Fra-1 and c-Fos share affinity for anionic lipids but the former has more affinity for a condensed phase and senses a change in DPPC phase, while the latter has more affinity for an expanded phase. These features are in contrast with our previous finding that c-Jun is not selective for phospholipid polar head group or charge. We show here that at least some immediate early transcription factors can interact with membrane phospholipids in a distinguishable manner, and this shall provide a basis for their potential capacity to regulate membrane-mediated cellular processes.


Subject(s)
Proto-Oncogene Proteins c-fos , Proto-Oncogene Proteins c-jun , Adsorption , Humans , Phospholipids/metabolism , Proto-Oncogene Proteins c-fos/chemistry , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism , Proto-Oncogene Proteins c-jun/chemistry , Proto-Oncogene Proteins c-jun/genetics , Proto-Oncogene Proteins c-jun/metabolism , Surface Properties
SELECTION OF CITATIONS
SEARCH DETAIL
...