Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
BMC Cancer ; 23(1): 613, 2023 Jul 03.
Article in English | MEDLINE | ID: mdl-37400764

ABSTRACT

BACKGROUND: Ovarian clear cell carcinomas (OCCCs) are rare, aggressive and chemoresistant tumors. Geographical and ethnic differences in the incidence of OCCC have been reported with a higher incidence in Asiatic countries. There is a paucity of information regarding OCCC in Latin America (LA) and other countries. METHODS: Here, we characterized two cohorts of 33 patients with OCCC from LA (24 from Brazil and 9 from Costa Rica) and a cohort of 27 patients from Spain. Genomic analysis was performed for 26 OCCC using the OncoScan platform. Tumors were classified according to their genomic landscapes into subgroups. Clinical parameters were related to the frequency of genomic aberrations. RESULTS: The median overall survival (OS) was not significantly different between the cohorts. Genomic landscapes were characterized by different homologous recombination deficiency (HRD) levels. No difference in the distribution of genomic landscapes profiles was detected between patients from the different cohorts. OCCCs with MYC-amplified tumors harboring a concomitant loss of a region in chromosome 13q12-q13 that includes the BRCA2 gene had the longest OS. In contrast, patients carrying a high number (> 30) of total copy number (CN) aberrations with no concomitant alterations in MYC and BRCA2 genes presented the shortest OS. Furthermore, amplification of the ASH1L gene was also associated with a shorter OS. Initial-stage OCCCs with early progression were characterized by gains in the JNK1 and MKL1 genes. CONCLUSIONS: Our results provide new data from understudied OCCC populations and reveal new potential markers for OCCCs.


Subject(s)
Adenocarcinoma, Clear Cell , Carcinoma , Ovarian Neoplasms , Female , Humans , Ovarian Neoplasms/pathology , Genomics , Brazil , Adenocarcinoma, Clear Cell/pathology
2.
Int J Mol Sci ; 24(21)2023 Nov 01.
Article in English | MEDLINE | ID: mdl-37958846

ABSTRACT

Glioblastoma (GBM) is the most common and aggressive primary brain tumor in adults, with few effective treatment strategies. The research on the development of new treatments is often constrained by the limitations of preclinical models, which fail to accurately replicate the disease's essential characteristics. Herein, we describe the obtention, molecular, and functional characterization of the GBM33 cell line. This cell line belongs to the GBM class according to the World Health Organization 2021 Classification of Central Nervous System Tumors, identified by methylation profiling. GBM33 expresses the astrocytic marker GFAP, as well as markers of neuronal origin commonly expressed in GBM cells, such as ßIII-tubulin and neurofilament. Functional assays demonstrated an increased growth rate when compared to the U87 commercial cell line and a similar sensitivity to temozolamide. GBM33 cells retained response to serum starvation, with reduced growth and diminished activation of the Akt signaling pathway. Unlike LN-18 and LN-229 commercial cell lines, GBM33 is able to produce primary cilia upon serum starvation. In summary, the successful establishment and comprehensive characterization of this GBM cell line provide researchers with invaluable tools for studying GBM biology, identifying novel therapeutic targets, and evaluating the efficacy of potential treatments.


Subject(s)
Brain Neoplasms , Glioblastoma , Adult , Humans , Glioblastoma/metabolism , Brazil , Brain Neoplasms/metabolism , Cell Line, Tumor , Tubulin/metabolism
3.
Nucleic Acids Res ; 46(1): e3, 2018 01 09.
Article in English | MEDLINE | ID: mdl-29069469

ABSTRACT

Polysome-profiling is commonly used to study translatomes and applies laborious extraction of efficiently translated mRNA (associated with >3 ribosomes) from a large volume across many fractions. This property makes polysome-profiling inconvenient for larger experimental designs or samples with low RNA amounts. To address this, we optimized a non-linear sucrose gradient which reproducibly enriches for efficiently translated mRNA in only one or two fractions, thereby reducing sample handling 5-10-fold. The technique generates polysome-associated RNA with a quality reflecting the starting material and, when coupled with smart-seq2 single-cell RNA sequencing, translatomes in small tissues from biobanks can be obtained. Translatomes acquired using optimized non-linear gradients resemble those obtained with the standard approach employing linear gradients. Polysome-profiling using optimized non-linear gradients in serum starved HCT-116 cells with or without p53 showed that p53 status associates with changes in mRNA abundance and translational efficiency leading to changes in protein levels. Moreover, p53 status also induced translational buffering whereby changes in mRNA levels are buffered at the level of mRNA translation. Thus, here we present a polysome-profiling technique applicable to large study designs, primary cells and frozen tissue samples such as those collected in biobanks.


Subject(s)
Polyribosomes/metabolism , Protein Biosynthesis , RNA, Messenger/genetics , Ribosomes/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Female , HCT116 Cells , Humans , MCF-7 Cells , Mutation , RNA, Messenger/metabolism , Sequence Analysis, RNA , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
4.
Int J Mol Sci ; 20(9)2019 May 02.
Article in English | MEDLINE | ID: mdl-31052505

ABSTRACT

Glioblastoma (GBM) is one of the most aggressive cancers, with median survival of less than 2 years. Despite of considerable advance in molecular classification of GBMs, no improvements in therapy have been described. The scenario is further complicated by tumor heterogeneity and the relationship among genetic, transcriptional and functional findings. Classically, gene expression has been evaluated by steady-state mRNA, however, this does not take translational control into consideration, which contributes considerably to the composition of the proteome. In this study, we evaluated the transcriptomic and translatomic signature of a GBM obtained from a single patient focusing in tumor heterogeneity. In a sampling of eight fragments, we investigated the translation rates, mTORC1 and ERK1/2 pathways and identified both total and polysome associated mRNAs. An increased translation rate was observed in fragments with high-grade histological features. High-grade histology was also associated with the expression of genes related to extracellular matrix (ECM) and angiogenesis, in both transcriptomes and translatomes. However, genes associated with epithelial to mesenchymal transition and stress response, were observed only in translatomes from high-grade fragments. Overall, our results demonstrate that isolation of translated mRNA can be used to identify biomarkers and reveal previously unrecognized determinants of heterogeneity in GBMs.


Subject(s)
Central Nervous System Neoplasms/genetics , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Glioblastoma/genetics , Cell Line, Tumor , Central Nervous System Neoplasms/pathology , Extracellular Signal-Regulated MAP Kinases/genetics , Female , Glioblastoma/pathology , Humans , Mechanistic Target of Rapamycin Complex 1/genetics , Middle Aged , Protein Biosynthesis , RNA, Messenger/genetics
5.
Biochem J ; 474(17): 2981-2991, 2017 08 17.
Article in English | MEDLINE | ID: mdl-28739602

ABSTRACT

Prion protein (PrPC) was initially described due to its involvement in transmissible spongiform encephalopathies. It was subsequently demonstrated to be a cell surface molecule involved in many physiological processes, such as vesicle trafficking. Here, we investigated the roles of PrPC in the response to insulin and obesity development. Two independent PrPC knockout (KO) and one PrPC overexpressing (TG20) mouse models were fed high-fat diets, and the development of insulin resistance and obesity was monitored. PrPC KO mice fed high-fat diets presented all of the symptoms associated with the development of insulin resistance: hyperglycemia, hyperinsulinemia, and obesity. Conversely, TG20 animals fed high-fat diets showed reduced weight and insulin resistance. Accordingly, the expression of peroxisome proliferator-activated receptor gamma (PPARγ) was reduced in PrPC KO mice and increased in TG20 animals. PrPC KO cells also presented reduced glucose uptake upon insulin stimulation, due to reduced translocation of the glucose transporter Glut4. Thus, our results suggest that PrPC reflects susceptibility to the development of insulin resistance and metabolic syndrome.


Subject(s)
Glucose Transporter Type 4/metabolism , Insulin Resistance , Obesity/metabolism , PPAR gamma/metabolism , PrPC Proteins/metabolism , Prion Proteins/metabolism , 3T3-L1 Cells , Animals , Cell Membrane/metabolism , Cell Membrane/pathology , Cells, Cultured , Crosses, Genetic , Diet, High-Fat/adverse effects , Embryo, Mammalian/pathology , Female , Gene Expression Regulation , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Obesity/etiology , Obesity/pathology , PPAR gamma/genetics , PrPC Proteins/genetics , Prion Proteins/genetics , Protein Transport , Weight Gain
6.
J Biol Chem ; 288(15): 10860-9, 2013 Apr 12.
Article in English | MEDLINE | ID: mdl-23447528

ABSTRACT

The product of the mouse Imprinted and Ancient gene, IMPACT, is preferentially expressed in neurons. We have previously shown that IMPACT overexpression inhibits the activation of the protein kinase GCN2, which signals amino acid starvation. GCN2 phosphorylates the α-subunit of eukaryotic translation initiation factor 2 (eIF2α), resulting in inhibition of general protein synthesis but increased translation of specific messages, such as ATF4. GCN2 is also involved in the regulation of neuronal functions, controlling synaptic plasticity, memory, and feeding behavior. We show here that IMPACT abundance increases during differentiation of neurons and neuron-like N2a cells, whereas GCN2 displays lowered activation levels. Upon differentiation, IMPACT associates with translating ribosomes, enhances translation initiation, and down-regulates the expression of ATF4. We further show that endogenous IMPACT promotes neurite outgrowth whereas GCN2 is a strong inhibitor of spontaneous neuritogenesis. Together, these results uncover the participation of the GCN2-IMPACT module of translational regulation in a highly controlled step in the development of the nervous system.


Subject(s)
Gene Expression Regulation, Developmental/physiology , Nerve Tissue Proteins/physiology , Neurites/metabolism , Neurogenesis/physiology , Protein Biosynthesis/physiology , Protein Serine-Threonine Kinases/metabolism , Proteins/metabolism , Activating Transcription Factor 4/biosynthesis , Activating Transcription Factor 4/genetics , Animals , Behavior, Animal/physiology , Cells, Cultured , Down-Regulation/physiology , Eukaryotic Initiation Factor-2/genetics , Eukaryotic Initiation Factor-2/metabolism , Feeding Behavior/physiology , Intracellular Signaling Peptides and Proteins , Memory/physiology , Mice , Mice, Knockout , Protein Serine-Threonine Kinases/genetics , Proteins/genetics , Ribosomes/genetics , Ribosomes/metabolism , Synapses/genetics , Synapses/metabolism
7.
FASEB J ; 27(9): 3594-607, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23729591

ABSTRACT

Stress-inducible phosphoprotein 1 (STI1) is part of the chaperone machinery, but it also functions as an extracellular ligand for the prion protein. However, the physiological relevance of these STI1 activities in vivo is unknown. Here, we show that in the absence of embryonic STI1, several Hsp90 client proteins are decreased by 50%, although Hsp90 levels are unaffected. Mutant STI1 mice showed increased caspase-3 activation and 50% impairment in cellular proliferation. Moreover, placental disruption and lack of cellular viability were linked to embryonic death by E10.5 in STI1-mutant mice. Rescue of embryonic lethality in these mutants, by transgenic expression of the STI1 gene, supported a unique role for STI1 during embryonic development. The response of STI1 haploinsufficient mice to cellular stress seemed compromised, and mutant mice showed increased vulnerability to ischemic insult. At the cellular level, ischemia increased the secretion of STI1 from wild-type astrocytes by 3-fold, whereas STI1 haploinsufficient mice secreted half as much STI1. Interesting, extracellular STI1 prevented ischemia-mediated neuronal death in a prion protein-dependent way. Our study reveals essential roles for intracellular and extracellular STI1 in cellular resilience.


Subject(s)
Embryo, Mammalian/metabolism , Heat-Shock Proteins/metabolism , Ischemia/metabolism , Molecular Chaperones/metabolism , Prions/metabolism , Animals , Blastocyst/metabolism , Blotting, Western , CDX2 Transcription Factor , Cells, Cultured , Female , Heat-Shock Proteins/genetics , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , In Vitro Techniques , Ischemia/genetics , Mice , Mice, Mutant Strains , Molecular Chaperones/genetics , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Pregnancy , Prions/genetics , Transcription Factors/genetics , Transcription Factors/metabolism
8.
Cell Mol Life Sci ; 70(17): 3211-27, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23543276

ABSTRACT

The co-chaperone stress-inducible protein 1 (STI1) is released by astrocytes, and has important neurotrophic properties upon binding to prion protein (PrP(C)). However, STI1 lacks a signal peptide and pharmacological approaches pointed that it does not follow a classical secretion mechanism. Ultracentrifugation, size exclusion chromatography, electron microscopy, vesicle labeling, and particle tracking analysis were used to identify three major types of extracellular vesicles (EVs) released from astrocytes with sizes ranging from 20-50, 100-200, and 300-400 nm. These EVs carry STI1 and present many exosomal markers, even though only a subpopulation had the typical exosomal morphology. The only protein, from those evaluated here, present exclusively in vesicles that have exosomal morphology was PrP(C). STI1 partially co-localized with Rab5 and Rab7 in endosomal compartments, and a dominant-negative for vacuolar protein sorting 4A (VPS4A), required for formation of multivesicular bodies (MVBs), impaired EV and STI1 release. Flow cytometry and PK digestion demonstrated that STI1 localized to the outer leaflet of EVs, and its association with EVs greatly increased STI1 activity upon PrP(C)-dependent neuronal signaling. These results indicate that astrocytes secrete a diverse population of EVs derived from MVBs that contain STI1 and suggest that the interaction between EVs and neuronal surface components enhances STI1-PrP(C) signaling.


Subject(s)
Carrier Proteins/metabolism , Heat-Shock Proteins/metabolism , Secretory Vesicles/metabolism , Animals , Astrocytes/cytology , Astrocytes/metabolism , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Hippocampus/cytology , Immunoblotting , Mice , PrPC Proteins/metabolism , Secretory Vesicles/ultrastructure
9.
J Biol Chem ; 287(52): 43777-88, 2012 Dec 21.
Article in English | MEDLINE | ID: mdl-23132868

ABSTRACT

Prions, the agents of transmissible spongiform encephalopathies, require the expression of prion protein (PrP(C)) to propagate disease. PrP(C) is converted into an abnormal insoluble form, PrP(Sc), that gains neurotoxic activity. Conversely, clinical manifestations of prion disease may occur either before or in the absence of PrP(Sc) deposits, but the loss of normal PrP(C) function contribution for the etiology of these diseases is still debatable. Prion disease-associated mutations in PrP(C) represent one of the best models to understand the impact of PrP(C) loss-of-function. PrP(C) associates with various molecules and, in particular, the interaction of PrP(C) with laminin (Ln) modulates neuronal plasticity and memory formation. To assess the functional alterations associated with PrP(C) mutations, wild-type and mutated PrP(C) proteins were expressed in a neural cell line derived from a PrP(C)-null mouse. Treatment with the laminin γ1 chain peptide (Ln γ1), which mimics the Ln binding site for PrP(C), increased intracellular calcium in cells expressing wild-type PrP(C), whereas a significantly lower response was observed in cells expressing mutated PrP(C) molecules. The Ln γ1 did not promote process outgrowth or protect against staurosporine-induced cell death in cells expressing mutated PrP(C) molecules in contrast to cells expressing wild-type PrP(C). The co-expression of wild-type PrP(C) with mutated PrP(C) molecules was able to rescue the Ln protective effects, indicating the lack of negative dominance of PrP(C) mutated molecules. These results indicate that PrP(C) mutations impair process outgrowth and survival mediated by Ln γ1 peptide in neural cells, which may contribute to the pathogenesis of genetic prion diseases.


Subject(s)
Laminin/metabolism , PrPC Proteins/metabolism , Animals , Binding Sites , Cell Line , Cell Survival/drug effects , Cell Survival/genetics , Enzyme Inhibitors/pharmacology , Genetic Diseases, Inborn/genetics , Genetic Diseases, Inborn/metabolism , Laminin/genetics , Mice , Mice, Mutant Strains , Mutation , PrPC Proteins/genetics , Prion Diseases/genetics , Prion Diseases/metabolism , Staurosporine/pharmacology
10.
J Neurochem ; 124(2): 210-23, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23145988

ABSTRACT

Prion protein (PrP(C)) is a cell surface glycoprotein that is abundantly expressed in nervous system. The elucidation of the PrP(C) interactome network and its significance on neural physiology is crucial to understanding neurodegenerative events associated with prion and Alzheimer's diseases. PrP(C) co-opts stress inducible protein 1/alpha7 nicotinic acetylcholine receptor (STI1/α7nAChR) or laminin/Type I metabotropic glutamate receptors (mGluR1/5) to modulate hippocampal neuronal survival and differentiation. However, potential cross-talk between these protein complexes and their role in peripheral neurons has never been addressed. To explore this issue, we investigated PrP(C)-mediated axonogenesis in peripheral neurons in response to STI1 and laminin-γ1 chain-derived peptide (Ln-γ1). STI1 and Ln-γ1 promoted robust axonogenesis in wild-type neurons, whereas no effect was observed in neurons from PrP(C) -null mice. PrP(C) binding to Ln-γ1 or STI1 led to an increase in intracellular Ca(2+) levels via distinct mechanisms: STI1 promoted extracellular Ca(2+) influx, and Ln-γ1 released calcium from intracellular stores. Both effects depend on phospholipase C activation, which is modulated by mGluR1/5 for Ln-γ1, but depends on, C-type transient receptor potential (TRPC) channels rather than α7nAChR for STI1. Treatment of neurons with suboptimal concentrations of both ligands led to synergistic actions on PrP(C)-mediated calcium response and axonogenesis. This effect was likely mediated by simultaneous binding of the two ligands to PrP(C). These results suggest a role for PrP(C) as an organizer of diverse multiprotein complexes, triggering specific signaling pathways and promoting axonogenesis in the peripheral nervous system.


Subject(s)
Calcium Signaling/physiology , Ganglia, Spinal/physiology , Heat-Shock Proteins/physiology , Laminin/physiology , PrPC Proteins/physiology , Receptor Cross-Talk/physiology , Sensory Receptor Cells/physiology , Animals , Axons/chemistry , Axons/physiology , Cell Survival/physiology , Extracellular Fluid/chemistry , Extracellular Fluid/physiology , Ganglia, Spinal/chemistry , Heat-Shock Proteins/chemistry , Intracellular Fluid/chemistry , Intracellular Fluid/metabolism , Laminin/metabolism , Mice , Mice, Knockout , Primary Cell Culture , Protein Binding/physiology , Sensory Receptor Cells/chemistry , Up-Regulation/physiology
11.
Proc Natl Acad Sci U S A ; 107(29): 13147-52, 2010 Jul 20.
Article in English | MEDLINE | ID: mdl-20615969

ABSTRACT

Transmissible spongiform encephalopathies are fatal neurodegenerative diseases caused by the conversion of prion protein (PrP(C)) into an infectious isoform (PrP(Sc)). How this event leads to pathology is not fully understood. Here we demonstrate that protein synthesis in neurons is enhanced via PrP(C) interaction with stress-inducible protein 1 (STI1). We also show that neuroprotection and neuritogenesis mediated by PrP(C)-STI1 engagement are dependent upon the increased protein synthesis mediated by PI3K-mTOR signaling. Strikingly, the translational stimulation mediated by PrP(C)-STI1 binding is corrupted in neuronal cell lines persistently infected with PrP(Sc), as well as in primary cultured hippocampal neurons acutely exposed to PrP(Sc). Consistent with this, high levels of eukaryotic translation initiation factor 2alpha (eIF2alpha) phosphorylation were found in PrP(Sc)-infected cells and in neurons acutely exposed to PrP(Sc). These data indicate that modulation of protein synthesis is critical for PrP(C)-STI1 neurotrophic functions, and point to the impairment of this process during PrP(Sc) infection as a possible contributor to neurodegeneration.


Subject(s)
Heat-Shock Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Neurons/metabolism , Prions/metabolism , Protein Biosynthesis , Protein Serine-Threonine Kinases/metabolism , Animals , Cell Line , Cytoprotection , Eukaryotic Initiation Factor-2/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , MAP Kinase Signaling System , Mice , Neurites/enzymology , Neurons/cytology , Neurons/enzymology , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , PrPSc Proteins/metabolism , Protein Binding , TOR Serine-Threonine Kinases , Up-Regulation
12.
FASEB J ; 25(1): 265-79, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20876210

ABSTRACT

The prion protein (PrP(C)) is highly expressed in the nervous system, and its abnormal conformer is associated with prion diseases. PrP(C) is anchored to cell membranes by glycosylphosphatidylinositol, and transmembrane proteins are likely required for PrP(C)-mediated intracellular signaling. Binding of laminin (Ln) to PrP(C) modulates neuronal plasticity and memory. We addressed signaling pathways triggered by PrP(C)-Ln interaction in order to identify transmembrane proteins involved in the transduction of PrP(C)-Ln signals. The Ln γ1-chain peptide, which contains the Ln binding site for PrP(C), induced neuritogenesis through activation of phospholipase C (PLC), Ca(2+) mobilization from intracellular stores, and protein kinase C and extracellular signal-regulated kinase (ERK1/2) activation in primary cultures of neurons from wild-type, but not PrP(C)-null mice. Phage display, coimmunoprecipitation, and colocalization experiments showed that group I metabotropic glutamate receptors (mGluR1/5) associate with PrP(C). Expression of either mGluR1 or mGluR5 in HEK293 cells reconstituted the signaling pathways mediated by PrP(C)-Ln γ1 peptide interaction. Specific inhibitors of these receptors impaired PrP(C)-Ln γ1 peptide-induced signaling and neuritogenesis. These data show that group I mGluRs are involved in the transduction of cellular signals triggered by PrP(C)-Ln, and they support the notion that PrP(C) participates in the assembly of multiprotein complexes with physiological functions on neurons.


Subject(s)
Laminin/metabolism , Neurites/physiology , PrPC Proteins/metabolism , Receptors, Metabotropic Glutamate/metabolism , Signal Transduction/physiology , Animals , Benzoates/pharmacology , Calcium/metabolism , Cells, Cultured , Female , Glycine/analogs & derivatives , Glycine/pharmacology , HEK293 Cells , Humans , Immunoblotting , Laminin/genetics , Laminin/pharmacology , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Neurites/metabolism , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , PrPC Proteins/genetics , Protein Binding , Pyridines/pharmacology , Receptor, Metabotropic Glutamate 5 , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Receptors, Metabotropic Glutamate/genetics , Type C Phospholipases/metabolism
13.
Cell Signal ; 79: 109883, 2021 03.
Article in English | MEDLINE | ID: mdl-33321181

ABSTRACT

Gliomas are the most frequent tumors of the central nervous system (CNS) and include the highly malignant glioblastoma (GBM). Characteristically, gliomas have translational control deregulation related to overactivation of signaling pathways such as PI3K/AKT/mTORC1 and Ras/ERK1/2. Thus, mRNA translation appears to play a dominant role in glioma gene expression patterns. The, analysis of genome-wide translated transcripts, together known as the translatome, may reveal important information for understanding gene expression patterns in gliomas. This review provides a brief overview of translational control mechanisms altered in gliomas with a focus on the current knowledge related to the translatomes of glioma cells and murine glioma models. We present an integrative meta-analysis of selected glioma translatome data with the aim of identifying recurrent patterns of gene expression preferentially regulated at the level of translation and obtaining clues regarding the pathological significance of these alterations. Re-analysis of several translatome datasets was performed to compare the translatomes of glioma models with those of their non-tumor counterparts and to document glioma cell responses to radiotherapy and MNK modulation. The role of recurrently altered genes in the context of translational control and tumorigenesis are discussed.


Subject(s)
Brain Neoplasms , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Glioma , Neoplasm Proteins , Protein Biosynthesis , Animals , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Genome-Wide Association Study , Glioma/genetics , Glioma/metabolism , Humans , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics
14.
Arch Oral Biol ; 118: 104865, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32801034

ABSTRACT

OBJECTIVE: The aim of this study was to verify ß2-AR expression in oral squamous cell carcinoma cell lines (SCC-9 and SCC-25), and to investigate the role of this receptor in migration and invasion of these neoplastic cells. DESIGN: SCC-9 and SCC-25 cells were investigated for gene and protein expression of ß2-AR. Cell migration and invasion were analyzed by wound healing assay and transwell invasion camera system. Different concentrations (0.1, 1 and 10 µM) of norepinephrine were used to stimulate, and 1 µM propranolol was used to block the beta-adrenergic receptors on cancer cells. Differences in median values of SCC-9 and SCC-25 and ß2-AR protein expression were analyzed by Friedman test and in case of significant differences; pairwise comparisons were performed using Bonferroni correction. RESULTS: The results showed that the ß2-AR gene and protein expression were observed in both oral cancer cell lines. The concentration of 10 µM of norepinephrine significantly inhibited (p ≤ 0.05) migration of SCC-9 and SCC-25 cell lines. Furthermore, there was a significant reduction (p ≤ 0.05) in the effect of norepinephrine on cell migration when the ß2-AR was inhibited by propranolol. The blockade by propranolol showed a tendency to reverse the effect of norepinephrine on the invasiveness of SCC-9 and SCC-25. CONCLUSIONS: The use of beta-adrenergic receptor agonists could become an adjuvant therapeutic target in the treatment of this malignancy.


Subject(s)
Carcinoma, Squamous Cell/pathology , Mouth Neoplasms/pathology , Neoplasm Invasiveness , Receptors, Adrenergic, beta/metabolism , Adrenergic beta-Antagonists/pharmacology , Carcinoma, Squamous Cell/drug therapy , Cell Line, Tumor , Humans , Mouth Neoplasms/drug therapy , Propranolol/pharmacology
15.
Mol Oncol ; 14(1): 159-179, 2020 01.
Article in English | MEDLINE | ID: mdl-31701625

ABSTRACT

The p90 ribosomal S6 kinase (RSK) family, a downstream target of Ras/extracellular signal-regulated kinase signaling, can mediate cross-talk with the mammalian target of rapamycin complex 1 pathway. As RSK connects two oncogenic pathways in gliomas, we investigated the protein levels of the RSK isoforms RSK1-4 in nontumoral brain (NB) and grade I-IV gliomas. When compared to NB or low-grade gliomas (LGG), a group of glioblastomas (GBMs) that excluded long-survivor cases expressed higher levels of RSK1 (RSK1hi ). No difference was observed in RSK2 median-expression levels among NB and gliomas; however, high levels of RSK2 in GBM (RSK2hi ) were associated with worse survival. RSK4 expression was not detected in any brain tissues, whereas RSK3 expression was very low, with GBM demonstrating the lowest RSK3 protein levels. RSK1hi and, to a lesser extent, RSK2hi GBMs showed higher levels of phosphorylated RSK, which reveals RSK activation. Transcriptome analysis indicated that most RSK1hi GBMs belonged to the mesenchymal subtype, and RSK1 expression strongly correlated with gene expression signature of immune infiltrates, in particular of activated natural killer cells and M2 macrophages. In an independent cohort, we confirmed that RSK1hi GBMs exclude long survivors, and RSK1 expression was associated with high protein levels of the mesenchymal subtype marker lysosomal protein transmembrane 5, as well as with high expression of CD68, which indicated the presence of infiltrating immune cells. An RSK1 signature was obtained based on differentially expressed mRNAs and validated in public glioma datasets. Enrichment of RSK1 signature followed glioma progression, recapitulating RSK1 protein expression, and was associated with worse survival not only in GBM but also in LGG. In conclusion, both RSK1 and RSK2 associate with glioma malignity, but displaying isoform-specific peculiarities. The progression-dependent expression and association with immune infiltration suggest RSK1 as a potential progression marker and therapeutic target for gliomas.


Subject(s)
Brain Neoplasms/metabolism , Glioma/metabolism , Lymphocytes, Tumor-Infiltrating/immunology , Membrane Proteins/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Transcriptome/immunology , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Brain Neoplasms/genetics , Brain Neoplasms/immunology , Brain Neoplasms/mortality , Databases, Genetic , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic/immunology , Glioblastoma/genetics , Glioblastoma/metabolism , Glioma/genetics , Glioma/immunology , Glioma/secondary , Humans , Immunohistochemistry , Killer Cells, Natural/metabolism , Macrophages/metabolism , Membrane Proteins/genetics , Neoplasm Grading , Phosphorylation , Protein Isoforms , Ribosomal Protein S6 Kinases, 90-kDa/genetics , Signal Transduction/genetics , Signal Transduction/immunology , Transcriptome/genetics
16.
Cancers (Basel) ; 12(5)2020 May 20.
Article in English | MEDLINE | ID: mdl-32443704

ABSTRACT

Multiple primary thyroid cancer (TC) and breast cancer (BC) are commonly diagnosed, and the lifetime risk for these cancers is increased in patients with a positive family history of both TC and BC. Although this phenotype is partially explained by TP53 or PTEN mutations, a significant number of patients are negative for these alterations. We judiciously recruited patients diagnosed with BC and/or TC having a family history of these tumors and assessed their whole-exome sequencing. After variant prioritization, we selected MUS81 c.1292G>A (p.R431H) for further investigation. This variant was genotyped in a healthy population and sporadic BC/TC tissues and investigated at the protein level and cellular models. MUS81 c.1292G>A was the most frequent variant (25%) and the strongest candidate due to its function of double-strand break repair. This variant was confirmed in four relatives from two families. MUS81 p.R431H protein exhibited lower expression levels in tumors from patients positive for the germline variant, compared with wild-type BC, and normal breast and thyroid tissues. Using cell line models, we showed that c.1292G>A induced protein instability and affected DNA damage response. We suggest that MUS81 is a novel candidate involved in familial BC/TC based on its low frequency in healthy individuals and proven effect in protein stability.

17.
Biochem Biophys Res Commun ; 378(1): 41-4, 2009 Jan 02.
Article in English | MEDLINE | ID: mdl-18996085

ABSTRACT

GCN2 is one of the four mammalian kinases that phosphorylate the alpha subunit of the translation initiation factor 2 (eIF2alpha) in a variety of stress situations, resulting in protein synthesis inhibition. GCN2 is involved in regulating metabolism, feeding behavior and memory in rodents. We show here that, relative to other cells, the beta isoform of the GCN2 transcript and the GCN2 protein are highly abundant in unfertilized mouse eggs. In addition, GCN2 in these cells is active, resulting in elevated levels of phosphorylated eIF2alpha. After fertilization, eIF2alpha phosphorylation decreases drastically. These results suggest that GCN2 mediated translational control may contribute to regulatory mechanisms operating during oocyte maturation.


Subject(s)
Eukaryotic Initiation Factor-2/metabolism , Oocytes/metabolism , Ovulation/metabolism , Protein Serine-Threonine Kinases/metabolism , Animals , Female , Mice , Mice, Inbred C57BL , Phosphorylation , Protein Serine-Threonine Kinases/genetics
18.
J Histochem Cytochem ; 66(6): 403-414, 2018 06.
Article in English | MEDLINE | ID: mdl-29328863

ABSTRACT

PI3K/Akt/mTOR pathway activation is a hallmark of high-grade gliomas, which prompted clinical trials for the use of PI3K and mTOR inhibitors. However, the poor results in the original trials suggested that better patient profiling was needed for such drugs. Thus, accurate and reproducible monitoring of mTOR complexes can lead to improved therapeutic strategies. In this work, we evaluated the expression and phosphorylation of mTOR, RAPTOR, and rpS6 in 195 human astrocytomas and 30 normal brain tissue samples. The expression of mTOR increased in glioblastomas, whereas mTOR phosphorylation, expression of RAPTOR, and expression and phosphorylation of rpS6 were similar between grades. Interestingly, the overexpression of total and phosphorylated mTOR as well as phosphorylated rpS6 (residues 240-244) were associated with wild-type IDH1 only glioblastomas. The expression and phosphorylation of mTOR and phosphorylation of rpS6 at residues 240-244 were associated with a worse prognosis in glioblastomas. Our results suggest that mTOR and rpS6 could be used as markers of overactivation of the PI3K-mTOR pathway and are predictive factors for overall survival in glioblastomas. Our study thus suggests that patients who harbor IDH1 wild-type glioblastomas might have increased benefit from targeted therapy against mTOR.


Subject(s)
Brain Neoplasms/pathology , Glioblastoma/pathology , Isocitrate Dehydrogenase/analysis , Ribosomal Protein S6 Kinases/analysis , TOR Serine-Threonine Kinases/analysis , Up-Regulation , Adult , Brain Neoplasms/diagnosis , Brain Neoplasms/epidemiology , Child , Female , Glioblastoma/diagnosis , Glioblastoma/epidemiology , Humans , Immunohistochemistry/methods , Male , Middle Aged , Phosphorylation , Prognosis , Survival Rate
19.
J Histochem Cytochem ; 65(2): 93-103, 2017 02.
Article in English | MEDLINE | ID: mdl-27789731

ABSTRACT

The mammalian target of rapamycin (mTOR) binds to several protein partners and forms two complexes, termed mTOR complexes 1 and 2 (mTORC1/2), that differ in components, substrates, and regulation. mTORC2 contains the protein Rapamycin-insensitive companion of mTOR (RICTOR); phosphorylates kinases of the AGC family, such as Akt; and controls the cytoskeleton. Even though the regulation of mTORC2 activity remains poorly understood, the hyperactivation of this signaling pathway has been shown to contribute to the oncogenic properties of gliomas in experimental models. In this work, we evaluated expression and phosphorylation of Akt, and expression of RICTOR and Ki-67 in 195 human astrocytomas of different grades (38 cases of grade I, 49 grade II, 15 grade III, and 93 grade IV) and 30 normal brains. Expression and phosphorylation of Akt increased with histological grade and correlated with a worse overall survival in glioblastomas (GBMs). RICTOR was overexpressed in grade I and II astrocytomas and demonstrated a shift to nuclear localization in GBMs. Nuclear RICTOR was associated to increased proliferation in GBMs. Our results point to an increase in total and phosphorylated Akt in high-grade gliomas and to a possible role of RICTOR in proliferations of high-grade GBM cells.


Subject(s)
Astrocytoma/pathology , Brain Neoplasms/pathology , Brain/pathology , Carrier Proteins/analysis , Proto-Oncogene Proteins c-akt/analysis , Adolescent , Adult , Aged , Cell Proliferation , Child , Child, Preschool , Female , Glioblastoma/pathology , Humans , Ki-67 Antigen/analysis , Male , Middle Aged , Phosphorylation , Rapamycin-Insensitive Companion of mTOR Protein , Young Adult
20.
Autophagy ; 12(11): 2113-2128, 2016 11.
Article in English | MEDLINE | ID: mdl-27629560

ABSTRACT

Prion protein modulates many cellular functions including the secretion of trophic factors by astrocytes. Some of these factors are found in exosomes, which are formed within multivesicular bodies (MVBs) and secreted into the extracellular space to modulate cell-cell communication. The mechanisms underlying exosome biogenesis were not completely deciphered. Here, we demonstrate that primary cultures of astrocytes and fibroblasts from prnp-null mice secreted lower levels of exosomes than wild-type cells. Furthermore, prnp-null astrocytes exhibited reduced MVB formation and increased autophagosome formation. The reconstitution of PRNP expression at the cell membrane restored exosome secretion in PRNP-deficient astrocytes, whereas macroautophagy/autophagy inhibition via BECN1 depletion reestablished exosome release in these cells. Moreover, the PRNP octapeptide repeat domain was necessary to promote exosome secretion and to impair the formation of the CAV1-dependent ATG12-ATG5 cytoplasmic complex that drives autophagosome formation. Accordingly, higher levels of CAV1 were found in lipid raft domains instead of in the cytoplasm in prnp-null cells. Collectively, these findings demonstrate that PRNP supports CAV1-suppressed autophagy to protect MVBs from sequestration into phagophores, thus facilitating exosome secretion.


Subject(s)
Autophagy , Caveolin 1/metabolism , Exosomes/metabolism , Prion Proteins/metabolism , Animals , Astrocytes/metabolism , Exosomes/ultrastructure , Lysosomes/metabolism , Membrane Microdomains/metabolism , Mice, Inbred C57BL , Models, Biological , Multivesicular Bodies/metabolism , Multivesicular Bodies/ultrastructure , Prion Proteins/chemistry , Protein Domains , Repetitive Sequences, Nucleic Acid , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL