Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 227
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Proc Natl Acad Sci U S A ; 118(39)2021 09 28.
Article in English | MEDLINE | ID: mdl-34561301

ABSTRACT

Nervous system malignancies are characterized by rapid progression and poor survival rates. These clinical observations underscore the need for novel therapeutic insights and pharmacological targets. To this end, here, we identify the orphan nuclear receptor NR5A2/LRH1 as a negative regulator of cancer cell proliferation and promising pharmacological target for nervous system-related tumors. In particular, clinical data from publicly available databases suggest that high expression levels of NR5A2 are associated with favorable prognosis in patients with glioblastoma and neuroblastoma tumors. Consistently, we experimentally show that NR5A2 is sufficient to strongly suppress proliferation of both human and mouse glioblastoma and neuroblastoma cells without inducing apoptosis. Moreover, short hairpin RNA-mediated knockdown of the basal expression levels of NR5A2 in glioblastoma cells promotes their cell cycle progression. The antiproliferative effect of NR5A2 is mediated by the transcriptional induction of negative regulators of the cell cycle, CDKN1A (encoding for p21cip1), CDKN1B (encoding for p27kip1) and Prox1 Interestingly, two well-established agonists of NR5A2, dilauroyl phosphatidylcholine (DLPC) and diundecanoyl phosphatidylcholine, are able to mimic the antiproliferative action of NR5A2 in human glioblastoma cells via the induction of the same critical genes. Most importantly, treatment with DLPC inhibits glioblastoma tumor growth in vivo in heterotopic and orthotopic xenograft mouse models. These data indicate a tumor suppressor role of NR5A2 in the nervous system and render this nuclear receptor a potential pharmacological target for the treatment of nervous tissue-related tumors.


Subject(s)
Glioblastoma/pathology , Nervous System Neoplasms/pathology , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , Cell Cycle/physiology , Cell Line, Tumor , Cell Proliferation , Glioblastoma/drug therapy , Glioblastoma/metabolism , Glioblastoma/mortality , Humans , Kaplan-Meier Estimate , Mice, SCID , Nervous System Neoplasms/drug therapy , Nervous System Neoplasms/metabolism , Nervous System Neoplasms/mortality , Neural Stem Cells/drug effects , Neuroblastoma/metabolism , Neuroblastoma/pathology , Phosphatidylcholines/pharmacology , Receptors, Cytoplasmic and Nuclear/agonists , Receptors, Cytoplasmic and Nuclear/genetics , Xenograft Model Antitumor Assays
2.
Int J Mol Sci ; 21(8)2020 Apr 20.
Article in English | MEDLINE | ID: mdl-32325928

ABSTRACT

Primary brain tumors, both malignant and benign, are diagnosed in adults at an incidence rate of approximately 23 people per 100 thousand. The role of AhR in carcinogenesis has been a subject of debate, given that this protein may act as either an oncogenic protein or a tumor suppressor in different cell types and contexts. Lately, there is growing evidence that aryl hydrocarbon receptor (AhR) plays an important part in the development of brain tumors. The role of AhR in brain tumors is complicated, depending on the type of tumor, on ligands that activate AhR, and other features of the pathological process. In this review, we summarize current knowledge about AhR in relation to brain tumors and provide an overview of AhR's potential as a therapeutic target.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Brain Neoplasms/etiology , Brain Neoplasms/metabolism , Disease Susceptibility , Receptors, Aryl Hydrocarbon/genetics , Receptors, Aryl Hydrocarbon/metabolism , Animals , Brain Neoplasms/pathology , Brain Neoplasms/therapy , Disease Management , Gene Expression Regulation, Neoplastic , Humans , Kynurenine/metabolism , Ligands , Metabolic Networks and Pathways , Molecular Targeted Therapy , Nervous System/metabolism , Nervous System/pathology , Nervous System Neoplasms/etiology , Nervous System Neoplasms/metabolism , Nervous System Neoplasms/pathology , Signal Transduction , Tryptophan/metabolism
3.
Biochem J ; 475(2): 531-545, 2018 01 31.
Article in English | MEDLINE | ID: mdl-29295892

ABSTRACT

The eukaryotic initiation factor 5A (eIF5A), which contributes to several crucial processes during protein translation, is the only protein that requires activation by a unique post-translational hypusine modification. eIF5A hypusination controls cell proliferation and has been linked to cancer. eIF5A hypusination requires the enzymes deoxyhypusine synthase (DHPS) and deoxyhypusine hydroxylase and uniquely depends on the polyamine (PA) spermidine as the sole substrate. Ornithine decarboxylase (ODC) is the rate-limiting enzyme in PA biosynthesis. Both ODC and PAs control cell proliferation and are frequently dysregulated in cancer. Since only spermidine can activate eIF5A, we chose the hypusine-PA nexus as a rational target to identify new drug combinations with synergistic antiproliferative effects. We show that elevated mRNA levels of the two target enzymes DHPS and ODC correlate with poor prognosis in a large cohort of neuroblastoma (NB) tumors. The DHPS inhibitor GC7 (N1-guanyl-1,7-diaminoheptane) and the ODC inhibitor α-difluoromethylornithine (DFMO) are target-specific and in combination induced synergistic effects in NB at concentrations that were not individually cytotoxic. Strikingly, while each drug alone at higher concentrations is known to induce p21/Rb- or p27/Rb-mediated G1 cell cycle arrest, we found that the drug combination induced caspase 3/7/9, but not caspase 8-mediated apoptosis, in NB cells. Hypusinated eIF5A levels and intracellular spermidine levels correlated directly with drug treatments, signifying specific drug targeting effects. This two-pronged GC7/DFMO combination approach specifically inhibits both spermidine biosynthesis and post-translational, spermidine-dependent hypusine-eIF5A activation, offering an exciting clue for improved NB drug therapy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols , Eflornithine/pharmacology , Gene Expression Regulation, Neoplastic , Guanine/analogs & derivatives , Nervous System Neoplasms/genetics , Neuroblastoma/genetics , Peptide Initiation Factors/genetics , RNA-Binding Proteins/genetics , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclin-Dependent Kinase Inhibitor p27/genetics , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Drug Synergism , Guanine/pharmacology , Humans , Lysine/analogs & derivatives , Lysine/metabolism , Mixed Function Oxygenases/genetics , Mixed Function Oxygenases/metabolism , Nervous System Neoplasms/metabolism , Nervous System Neoplasms/mortality , Nervous System Neoplasms/pathology , Neuroblastoma/metabolism , Neuroblastoma/mortality , Neuroblastoma/pathology , Ornithine Decarboxylase/genetics , Ornithine Decarboxylase/metabolism , Oxidoreductases Acting on CH-NH Group Donors/genetics , Oxidoreductases Acting on CH-NH Group Donors/metabolism , Peptide Initiation Factors/antagonists & inhibitors , Peptide Initiation Factors/metabolism , Prognosis , Protein Processing, Post-Translational , RNA-Binding Proteins/antagonists & inhibitors , RNA-Binding Proteins/metabolism , Retinoblastoma Protein/genetics , Retinoblastoma Protein/metabolism , Signal Transduction , Spermidine/metabolism , Eukaryotic Translation Initiation Factor 5A
4.
Genet Mol Res ; 16(1)2017 Mar 30.
Article in English | MEDLINE | ID: mdl-28362995

ABSTRACT

MiR-200b, a member of the microRNA-200 family, has been identified to be capable of suppressing glioma cell growth through targeting CREB1 or CD133. However, whether miR-200b affects the biological behavior (proliferation, invasion, and migration) of glioma cells is poorly understood. The aim of this study was to evaluate the effect of miR-200b on the biological behavior of glioma cells in vitro. MiRNA-200b mimics, miRNA-200b inhibitor, and mimic control were transfected into conventionally cultured glioma U251 cells, followed by measuring the expression of miR-200b and CD133 in transfected cells by RT-PCR; effect of miR-200b on CD133 mRNA 3'-UTR luciferase activity by luciferase reporter assay; proliferation activity of transfected U251 cells by MTT method; and changes in U251 cell invasion and migration by Transwell method after transfection. Compared to that in the miRNA-200b inhibitor, mimic control, and blank control groups, miRNA-200b expression was significantly increased and CD133 mRNA expression was significantly decreased in the mimic miRNA-200b group in a time-dependent manner (P < 0.05). Meanwhile, dual luciferase reporter assay showed that miR-200b could inhibit CD133 activity through binding to the 3'-UTR of CD133 mRNA (P < 0.05). Furthermore, the proliferation activity and invasion and migration abilities of U251 cells transfected with miRNA-200b mimic were significantly decreased (P < 0.05). In conclusion, overexpression of miR-200b inhibited the proliferation, invasion, and migration of glioma cells possibly through targeting CD133.


Subject(s)
AC133 Antigen/genetics , Glioma/genetics , MicroRNAs/genetics , 3' Untranslated Regions , AC133 Antigen/metabolism , Apoptosis/genetics , Cell Cycle/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , Glioma/metabolism , Glioma/pathology , Humans , MicroRNAs/biosynthesis , MicroRNAs/metabolism , Nervous System Neoplasms/genetics , Nervous System Neoplasms/metabolism , Nervous System Neoplasms/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transfection
5.
Hum Mol Genet ; 23(25): 6826-37, 2014 Dec 20.
Article in English | MEDLINE | ID: mdl-25104850

ABSTRACT

Uncontrolled cell cycle entry, resulting from deregulated CDK-RB1-E2F pathway activity, is a crucial determinant of neuroblastoma cell malignancy. Here we identify neuroblastoma-suppressive functions of the p19-INK4d CDK inhibitor and uncover mechanisms of its repression in high-risk neuroblastomas. Reduced p19-INK4d expression was associated with poor event-free and overall survival and neuroblastoma risk factors including amplified MYCN in a set of 478 primary neuroblastomas. High MYCN expression repressed p19-INK4d mRNA and protein levels in different neuroblastoma cell models with conditional MYCN expression. MassARRAY and 450K methylation analyses of 105 primary neuroblastomas uncovered a differentially methylated region within p19-INK4d. Hypermethylation of this region was associated with reduced p19-INK4d expression. In accordance, p19-INK4d expression was activated upon treatment with the demethylating agent, 2'-deoxy-5-azacytidine, in neuroblastoma cell lines. Ectopic p19-INK4d expression decreased viability, clonogenicity and the capacity for anchorage-independent growth of neuroblastoma cells, and shifted the cell cycle towards the G1/0 phase. p19-INK4d also induced neurite-like processes and markers of neuronal differentiation. Moreover, neuroblastoma cell differentiation, induced by all-trans retinoic acid or NGF-NTRK1-signaling, activated p19-INK4d expression. Our findings pinpoint p19-INK4d as a neuroblastoma suppressor and provide evidence for MYCN-mediated repression and for epigenetic silencing of p19-INK4d by DNA hypermethylation in high-risk neuroblastomas.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p19/genetics , Gene Expression Regulation, Neoplastic , Nervous System Neoplasms/genetics , Neuroblastoma/genetics , Neurons/metabolism , Nuclear Proteins/genetics , Oncogene Proteins/genetics , Adolescent , Adult , Antimetabolites, Antineoplastic/pharmacology , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Cell Differentiation/drug effects , Cell Line, Tumor , Child , Child, Preschool , Cyclin-Dependent Kinase Inhibitor p19/metabolism , DNA Methylation/drug effects , Decitabine , Epigenesis, Genetic , Female , Humans , Infant , Infant, Newborn , Male , N-Myc Proto-Oncogene Protein , Neoplasm Staging , Nervous System Neoplasms/metabolism , Nervous System Neoplasms/mortality , Nervous System Neoplasms/pathology , Neuroblastoma/metabolism , Neuroblastoma/mortality , Neuroblastoma/pathology , Neurons/drug effects , Neurons/pathology , Nuclear Proteins/metabolism , Oncogene Proteins/metabolism , Signal Transduction , Survival Analysis , Tretinoin/pharmacology
6.
Genes Chromosomes Cancer ; 52(2): 191-201, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23097141

ABSTRACT

Copy number changes or reduced expression of the Neuron navigator 3 (NAV3) gene occurs in neuroblastomas and malignancies of epithelial or lymphoid origin. To elucidate whether NAV3 has a role in the tumorigenesis of nervous system tumors in general, we studied central and peripheral nervous system tumors for NAV3 copy number changes. In search for common tumorigenic denominators, we analyzed 113 central and peripheral nervous system tumors, including glial tumors (grades I-IV gliomas), medulloblastomas, and neuroblastomas. NAV3 copy number changes were studied by fluorescence in situ hybridization and correlated to survival analyses. To identify target genes of NAV3 deletion, NAV3 was silenced by siRNA in glioblastoma cell lines and gene expression profiles were analyzed by Agilent 4×44k dual-color microarrays. Selected upregulations were confirmed by immunohistochemistry and quantitative polymerase chain reaction. We found NAV3 amplifications to dominate in neuronally differentiated tumors, whereas glial tumors showed almost equal proportions of NAV3 deletion and amplification. However, Grade IV gliomas had more frequent NAV3 deletions than grades I-III gliomas. Silencing of NAV3 in glioma cell lines led to the upregulation of receptor genes associated with gonadotropin-releasing hormone and Jak-Stat signaling pathways. Kaplan-Meier analysis of the entire clinical tumor material showed association between NAV3 amplifications and favorable prognosis, as well as NAV3 deletions and unfavorable prognosis. With Cox regression model, a hazard ratio of 0.51 was observed for NAV3 amplifications and 1.36 for NAV3 deletions. We conclude that NAV3 may be a potential new prognostic biomarker and a potential therapeutic target.


Subject(s)
DNA Copy Number Variations , Glioma/genetics , Medulloblastoma/genetics , Membrane Proteins/genetics , Nerve Tissue Proteins/genetics , Nervous System Neoplasms/genetics , Neuroblastoma/genetics , Biomarkers, Tumor/genetics , Cell Line, Tumor , Comparative Genomic Hybridization , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Glioma/metabolism , Glioma/pathology , Humans , Immunohistochemistry/statistics & numerical data , In Situ Hybridization, Fluorescence , Kaplan-Meier Estimate , Medulloblastoma/metabolism , Medulloblastoma/pathology , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Nervous System Neoplasms/metabolism , Nervous System Neoplasms/pathology , Neuroblastoma/metabolism , Neuroblastoma/pathology , Oligonucleotide Array Sequence Analysis/methods , Oligonucleotide Array Sequence Analysis/statistics & numerical data , Prognosis , Proportional Hazards Models , RNA Interference , Receptors, Interleukin/genetics , Receptors, Interleukin/metabolism , Receptors, LHRH/genetics , Receptors, LHRH/metabolism , Reverse Transcriptase Polymerase Chain Reaction
7.
Cells ; 13(8)2024 Apr 11.
Article in English | MEDLINE | ID: mdl-38667282

ABSTRACT

Transglutaminase type 2 (TG2) is the most ubiquitously expressed member of the transglutaminase family. TG2 catalyzes the transamidation reaction leading to several protein post-translational modifications and it is also implicated in signal transduction thanks to its GTP binding/hydrolyzing activity. In the nervous system, TG2 regulates multiple physiological processes, such as development, neuronal cell death and differentiation, and synaptic plasticity. Given its different enzymatic activities, aberrant expression or activity of TG2 can contribute to tumorigenesis, including in peripheral and central nervous system tumors. Indeed, TG2 dysregulation has been reported in meningiomas, medulloblastomas, neuroblastomas, glioblastomas, and other adult-type diffuse gliomas. The aim of this review is to provide an overview of the biological and functional relevance of TG2 in the pathogenesis of nervous system tumors, highlighting its involvement in survival, tumor inflammation, differentiation, and in the resistance to standard therapies.


Subject(s)
GTP-Binding Proteins , Nervous System Neoplasms , Protein Glutamine gamma Glutamyltransferase 2 , Animals , Humans , GTP-Binding Proteins/metabolism , Nervous System Neoplasms/pathology , Nervous System Neoplasms/enzymology , Nervous System Neoplasms/metabolism , Transglutaminases/metabolism
8.
Am J Pathol ; 180(3): 1232-1242, 2012 03.
Article in English | MEDLINE | ID: mdl-22252234

ABSTRACT

Perineural invasion (PNI) is a tropism of tumor cells for nerve bundles located in the surrounding stroma. It is a pathological feature observed in certain tumors, referred to as neurotropic malignancies, that severely limits the ability to establish local control of disease and results in pain, recurrent growth, and distant metastases. Despite the importance of PNI as a prognostic indicator, its biological mechanisms are poorly understood. The semaphorins and their receptors, the plexins, compose a family of proteins originally shown to be important in nerve cell adhesion, axon migration, and proper central nervous system development. Emerging evidence has demonstrated that these factors are expressed in tissues outside of the nervous system and represent a widespread signal transduction system that is involved in the regulation of motility and adhesion in different cell types. We believe that the plexins and semaphorins, which are strongly expressed in both axons and many carcinomas, play a role in PNI. In this study, we show that plexin-B1 is overexpressed in tissues and cell lines from neurotropic malignancies and is attracted to nerves that express its ligand, semaphorin 4D, in a Rho/Rho kinase-dependent manner. We also demonstrate that nerves are attracted to tumors through this same system of proteins, suggesting that both plexin-B1 and semaphorin 4D are important in the promotion of PNI.


Subject(s)
Antigens, CD/physiology , Nerve Tissue Proteins/physiology , Nervous System Neoplasms/pathology , Receptors, Cell Surface/physiology , Semaphorins/physiology , rhoA GTP-Binding Protein/metabolism , Animals , Antigens, CD/metabolism , Axons/physiology , Cell Line, Tumor , Cell Movement/physiology , Drug Synergism , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Nude , Neoplasm Invasiveness , Neoplasm Transplantation , Nerve Tissue Proteins/metabolism , Nervous System Neoplasms/metabolism , RNA, Small Interfering/pharmacology , Receptors, Cell Surface/metabolism , Semaphorins/metabolism , Signal Transduction , Transplantation, Heterologous
9.
BMC Cancer ; 13: 35, 2013 Jan 26.
Article in English | MEDLINE | ID: mdl-23351358

ABSTRACT

BACKGROUND: Changes in cell shape and plasticity in cytoskeletal dynamics are critically involved in cell adhesion, migration, invasion and the overall process of metastasis. Previous work in our laboratory demonstrated that the synthetic steroid mifepristone inhibited the growth of highly metastatic cancer cells, while simultaneously causing striking changes in cellular morphology. Here we assessed whether such morphological alterations developed in response to cytostatic concentrations of mifepristone are reversible or permanent, involve rearrangement of cytoskeletal proteins, and/or affect the adhesive capacity of the cells. METHODS: Cancer cell lines of the ovary (SKOV-3), breast (MDA-MB-231), prostate (LNCaP), and nervous system (U87MG) were exposed to cytostatic concentrations of mifepristone and studied by phase-contrast microscopy. The transient or permanent nature of the cytostasis and morphological changes caused by mifepristone was assessed, as well as the rearrangement of cytoskeletal proteins. De-adhesion and adhesion assays were utilized to determine if mifepristone-arrested and morphologically dysregulated cells had abnormal de-adhesion/adhesion dynamics when compared to vehicle-treated controls. RESULTS: Mifepristone-treated cells displayed a long, thin, spindle-like shape with boundaries resembling those of loosely adhered cells. Growth arrest and morphology changes caused by mifepristone were reversible in SKOV-3, MDA-MB-231 and U87MG, but not in LNCaP cells that instead became senescent. All cancer cell types exposed to mifepristone displayed greatly increased actin ruffling in association with accelerated de-adhesion from the culture plate, and delayed adhesion capacity to various extracellular matrix components. CONCLUSIONS: Cytostatic concentrations of mifepristone induced alterations in the cellular structure of a panel of aggressive, highly metastatic cancer cells of different tissues of origin. Such changes were associated with re-distribution of actin fibers that mainly form non-adhesive membrane ruffles, leading to dysregulated cellular adhesion capacity.


Subject(s)
Actin Cytoskeleton/drug effects , Antineoplastic Agents, Hormonal/pharmacology , Cell Adhesion/drug effects , Cell Proliferation/drug effects , Cell Shape/drug effects , Cytostatic Agents/pharmacology , Hormone Antagonists/pharmacology , Mifepristone/pharmacology , Neoplasms/pathology , Actin Cytoskeleton/metabolism , Actin Cytoskeleton/pathology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Humans , Male , Neoplasm Invasiveness , Neoplasms/metabolism , Nervous System Neoplasms/metabolism , Nervous System Neoplasms/pathology , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Time Factors
10.
Genes Chromosomes Cancer ; 51(2): 174-85, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22045684

ABSTRACT

Epigenetic modifications such as methylation of CpG islands in tumor-suppressor gene promoter regions have been associated with tumor development in many human cancers. Using methylation specific multiplex ligation-dependent probe amplification method, we analyzed the methylation status of 35 different genes in 16 neuroblastoma (NB) cell lines and 50 NB tumor samples (NBs), and investigated whether specific hypermethylation was associated with biological and/or clinical parameters. Among the genes found hypermethylated, the effect of GSTP1 hypermethylation on mRNA and protein expression was also explored. The median number of hypermethylated genes was higher in cell lines compared to NBs (5.5 vs. 2). For eight genes, aberrant methylation of CpG-islands in NB was not (ESR1, PAX5, WT1, CADM1, MSH6, and CDKN2B) or very rarely (CDH13 and GSTP1) reported in literature. GSTP1 was found hypermethylated in 44% of the NB cell lines and in 33% of the stage 4-11qLOH -non MYCN-amplified high risk NBs. Hypermethylation was correlated with reduced mRNA and protein expression. In the whole NBs cohort, GSTP1 hypermethylation was less frequently detected (8%), but found to be associated with lower event-free (EFS) and overall survival. Hypermethylation of GSTP1 showed also association with lower EFS in high risk subgroups as stage 4 and older patients (≥547 days). Our results suggest that, as in several adult cancers, aberrant methylation of GSTP1 may contribute to the carcinogenetic process in NB and could be potentially used as a new marker leading to define an ultra-high risk subgroup.


Subject(s)
Biomarkers, Tumor/genetics , DNA Methylation , Down-Regulation , Glutathione S-Transferase pi/genetics , Nervous System Neoplasms/genetics , Neuroblastoma/genetics , Adolescent , Biomarkers, Tumor/metabolism , Case-Control Studies , Cell Line, Tumor , Child , Child, Preschool , Gene Expression , Gene Expression Regulation, Neoplastic , Genetic Association Studies , Glutathione S-Transferase pi/metabolism , Humans , Infant , Infant, Newborn , Kaplan-Meier Estimate , Loss of Heterozygosity , Nervous System Neoplasms/metabolism , Nervous System Neoplasms/mortality , Nervous System Neoplasms/pathology , Neuroblastoma/metabolism , Neuroblastoma/mortality , Neuroblastoma/pathology , Prognosis , Promoter Regions, Genetic
11.
Dev Cell ; 58(2): 81-93, 2023 01 23.
Article in English | MEDLINE | ID: mdl-36693322

ABSTRACT

Similar to their pivotal roles in nervous system development, neurons have emerged as critical regulators of cancer initiation, maintenance, and progression. Focusing on nervous system tumors, we describe the normal relationships between neurons and other cell types relevant to normal nerve function, and discuss how disruptions of these interactions promote tumor evolution, focusing on electrical (gap junctions) and chemical (synaptic) coupling, as well as the establishment of new paracrine relationships. We also review how neuron-tumor communication contributes to some of the complications of cancer, including neuropathy, chemobrain, seizures, and pain. Finally, we consider the implications of cancer neuroscience in establishing risk for tumor penetrance and in the design of future anti-tumoral treatments.


Subject(s)
Nervous System Neoplasms , Neurons , Humans , Neurons/metabolism , Gap Junctions/metabolism , Nervous System Neoplasms/metabolism
12.
Life Sci ; 335: 122275, 2023 Dec 15.
Article in English | MEDLINE | ID: mdl-37984514

ABSTRACT

Cancer and stem cells share many characteristics related to self-renewal and differentiation. Both cell types express the same critical proteins that govern cellular stemness, which provide cancer cells with the growth and survival benefits of stem cells. LIN28 is an example of one such protein. LIN28 includes two main isoforms, LIN28A and LIN28B, with diverse physiological functions from tissue development to control of pluripotency. In addition to their physiological roles, LIN28A and LIN28B affect the progression of several cancers by regulating multiple cancer hallmarks. Altered expression levels of LIN28A and LIN28B have been proposed as diagnostic and/or prognostic markers for various malignancies. This review discusses the structure and modes of action of the different LIN28 proteins and examines their roles in regulating cancer hallmarks with a focus on malignancies of the nervous system. This review also highlights some gaps in the field that require further exploration to assess the potential of targeting LIN28 proteins for controlling cancer.


Subject(s)
MicroRNAs , Neoplasms , Nervous System Neoplasms , Humans , Neoplasms/metabolism , Nervous System Neoplasms/metabolism , Stem Cells/metabolism , DNA-Binding Proteins/metabolism , MicroRNAs/metabolism
13.
Mol Syst Biol ; 7: 486, 2011 Apr 26.
Article in English | MEDLINE | ID: mdl-21525872

ABSTRACT

DNA copy number aberrations (CNAs) are a hallmark of cancer genomes. However, little is known about how such changes affect global gene expression. We develop a modeling framework, EPoC (Endogenous Perturbation analysis of Cancer), to (1) detect disease-driving CNAs and their effect on target mRNA expression, and to (2) stratify cancer patients into long- and short-term survivors. Our method constructs causal network models of gene expression by combining genome-wide DNA- and RNA-level data. Prognostic scores are obtained from a singular value decomposition of the networks. By applying EPoC to glioblastoma data from The Cancer Genome Atlas consortium, we demonstrate that the resulting network models contain known disease-relevant hub genes, reveal interesting candidate hubs, and uncover predictors of patient survival. Targeted validations in four glioblastoma cell lines support selected predictions, and implicate the p53-interacting protein Necdin in suppressing glioblastoma cell growth. We conclude that large-scale network modeling of the effects of CNAs on gene expression may provide insights into the biology of human cancer. Free software in MATLAB and R is provided.


Subject(s)
Gene Dosage , Glioblastoma/genetics , Nerve Tissue Proteins/metabolism , Nervous System Neoplasms/genetics , Nuclear Proteins/metabolism , Transcriptional Activation/genetics , Tumor Suppressor Protein p53/metabolism , Cell Line, Tumor , Chromosome Aberrations , Databases, Factual , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Genome, Human , Genome-Wide Association Study , Glioblastoma/metabolism , Glioblastoma/mortality , Glioblastoma/pathology , Humans , Models, Genetic , Nerve Tissue Proteins/genetics , Nervous System Neoplasms/metabolism , Nervous System Neoplasms/mortality , Nervous System Neoplasms/pathology , Nuclear Proteins/genetics , Prognosis , Software , Tumor Suppressor Protein p53/genetics
14.
J Surg Res ; 173(2): 314-9, 2012 Apr.
Article in English | MEDLINE | ID: mdl-21195422

ABSTRACT

BACKGROUND: L1 cell adhesion molecule (CD171) has been detected in different malignant tumors and is associated with unfavorable outcome. It thus represents a target for tumor diagnosis and therapy. In this study, we assessed L1 expression in more than 8000 normal human tissues and different types of tumors, both malignant and non-malignant, and neural and non-neural. MATERIALS AND METHODS: Tissue micro-arrays, including a multi-tumor-array of 128 different tumor types, up to 50 samples of each type (approximately 5500 different samples), arrays with approximately 3000 different prostate and 600 mesenchymal tumor samples, and a normal human tissue-array were analyzed by immunohistochemistry with a monoclonal antibody using immunoperoxidase staining. RESULTS: L1 expression was detected in tumors of neural and neural crest origin and other types of non-neural tumors, but not in those of epithelial origin. In normal human tissues, L1 was detected in skin basal cells and small blood vessels, most notably in the mature placenta and peripheral nerves. CONCLUSION: This first comprehensive study of the importance of L1 expression in human demonstrates strong L1 overexpression in tumors of neuroectodermal and neural crest origin and an expression in only very few normal human tissues. L1 thus is a potentially important therapeutic target, particularly with respect to malignant melanoma, gastrointestinal stromal tumor, neuroblastoma, and certain subtypes of non-neural tumors.


Subject(s)
Biomarkers, Tumor/metabolism , Nervous System Neoplasms/metabolism , Neural Cell Adhesion Molecule L1/metabolism , Humans , Tissue Array Analysis
15.
J Pediatr Endocrinol Metab ; 25(5-6): 453-8, 2012.
Article in English | MEDLINE | ID: mdl-22876538

ABSTRACT

OBJECTIVE: We assessed the gonadal function in boys with a newly diagnosed neoplastic disease prior to chemotherapy. Eighty-four boys (48 prepubertal and 36 pubertal) were evaluated, including 50 with acute lymphoblastic leukemia (ALL) or non-Hodgkin lymphoma (NHL), 10 with Hodgkin lymphoma (HL), and 24 with solid tumors. The control group consisted of 24 healthy prepubertal and 24 pubertal boys. The levels of follicle-stimulating hormone (FSH), luteinizing hormone (LH), inhibin B, and testosterone were determined, and testicular volumes were measured. RESULTS: Patients in prepuberty and early puberty (Tanner stages 1-3) diagnosed with ALL/NHL or solid tumor presented normal serum reproductive hormone levels, whereas in ALL/NHL patients in Tanner stages 4-5, the mean values of inhibin B were significantly lower (45.18 +/- 33.85 vs. 153.57 +/- 71.44 ng/L, p = 0.0027). In patients with HL in Tanner stages 4-5, a statistically significant lower mean inhibin B level (100.44 +/- 67.45 versus 153.57 +/-71.44 ng/L, p = 0.0027), higher mean FSH level (6.3 +/- 3.6 versus 4.6 +/- 2.2 mIU/mL, p = 0.05), and higher mean LH level (5.9 +/- 4.0 versus 3.6 +/- 1.8 mIU/mL, p = 0.05) were observed. No statistically significant differences were noted in assessed hormones in patients with solid tumors, independently of Tanner stage. CONCLUSION: Our analysis indicates that adolescents with ALL/NHL and HL prior to treatment, exhibit reduced levels of inhibin B, which indirectly suggests the possibility of spermatogenesis dysfunction.


Subject(s)
Hormones/blood , Neoplasms , Puberty/physiology , Testis/physiology , Adolescent , Bone Neoplasms/diagnosis , Bone Neoplasms/metabolism , Bone Neoplasms/physiopathology , Child , Child, Preschool , Follicle Stimulating Hormone/blood , Hodgkin Disease/diagnosis , Hodgkin Disease/metabolism , Hodgkin Disease/physiopathology , Humans , Inhibins/blood , Kidney Neoplasms/diagnosis , Kidney Neoplasms/metabolism , Kidney Neoplasms/physiopathology , Luteinizing Hormone/blood , Lymphoma, Non-Hodgkin/diagnosis , Lymphoma, Non-Hodgkin/metabolism , Lymphoma, Non-Hodgkin/physiopathology , Male , Neoplasms/diagnosis , Neoplasms/metabolism , Neoplasms/physiopathology , Nervous System Neoplasms/diagnosis , Nervous System Neoplasms/metabolism , Nervous System Neoplasms/physiopathology , Neuroblastoma/diagnosis , Neuroblastoma/metabolism , Neuroblastoma/physiopathology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/diagnosis , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/physiopathology , Sarcoma/diagnosis , Sarcoma/metabolism , Sarcoma/physiopathology , Soft Tissue Neoplasms/diagnosis , Soft Tissue Neoplasms/metabolism , Soft Tissue Neoplasms/physiopathology , Testosterone/blood , Wilms Tumor/diagnosis , Wilms Tumor/metabolism , Wilms Tumor/physiopathology
16.
Cell Mol Life Sci ; 67(19): 3219-40, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20556632

ABSTRACT

Cell migration is essential in a number of processes, including wound healing, angiogenesis and cancer metastasis. Especially, invasion of cancer cells in the surrounding tissue is a crucial step that requires increased cell motility. Cell migration is a well-orchestrated process that involves the continuous formation and disassembly of matrix adhesions. Those structural anchor points interact with the extra-cellular matrix and also participate in adhesion-dependent signalling. Although these processes are essential for cancer metastasis, little is known about the molecular mechanisms that regulate adhesion dynamics during tumour cell migration. In this review, we provide an overview of recent advanced imaging strategies together with quantitative image analysis that can be implemented to understand the dynamics of matrix adhesions and its molecular components in relation to tumour cell migration. This dynamic cell imaging together with multiparametric image analysis will help in understanding the molecular mechanisms that define cancer cell migration.


Subject(s)
Cell Movement/physiology , Neoplasms/pathology , Animals , Cellular Structures/pathology , Cytoskeleton/metabolism , Cytoskeleton/pathology , Humans , Microscopy , Neoplasms/metabolism , Nervous System Neoplasms/metabolism , Signal Transduction , Wound Healing
17.
Proc Natl Acad Sci U S A ; 105(30): 10408-13, 2008 Jul 29.
Article in English | MEDLINE | ID: mdl-18641120

ABSTRACT

Zebrafish carrying heterozygous mutations for 17 different ribosomal protein (rp) genes are prone to developing malignant peripheral nerve sheath tumors (MPNSTs), a tumor type that is seldom seen in laboratory strains of zebrafish. Interestingly, the same rare tumor type arises in zebrafish that are homozygous for a loss-of-function point mutation in the tumor suppressor gene p53. For these reasons, and because p53 is widely known to be mutated in the majority of human cancers, we investigated the status of p53 in the rp(+/-) MPNSTs. Using monoclonal antibodies that we raised to zebrafish p53, we found that cells derived from rp(+/-) MPNSTs are significantly impaired in their ability to produce p53 protein even in the presence of a proteasome inhibitor and gamma-irradiation. Although the coding regions of the p53 gene remain wild type, the gene is transcribed, and overall protein production rates appear normal in rp(+/-) MPNST cells, p53 protein does not get synthesized. This defect is observed in all MPNSTs we examined that were derived from our 17 zebrafish lines with rp gene mutations. To date, studies of p53 in malignancies have focused predominantly on either p53 gene mutations or the aberrant posttranslational regulation of the p53 protein. Our results show that the appropriate amount of numerous ribosomal proteins is required for p53 protein production in vivo and that disruption of this regulation most likely contributes to tumorigenesis.


Subject(s)
Gene Expression Regulation, Neoplastic , Nerve Sheath Neoplasms/genetics , Nervous System Neoplasms/genetics , Point Mutation , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/physiology , Animals , Cell Line , Humans , Models, Genetic , Nerve Sheath Neoplasms/metabolism , Nervous System Neoplasms/metabolism , Oligonucleotide Array Sequence Analysis , Proteasome Inhibitors , Protein Processing, Post-Translational , Ribosomal Proteins/chemistry , Sequence Analysis, DNA , Zebrafish
18.
Cells ; 10(3)2021 03 06.
Article in English | MEDLINE | ID: mdl-33800887

ABSTRACT

For nearly a decade, researchers in the field of pediatric oncology have been using zebrafish as a model for understanding the contributions of genetic alternations to the pathogenesis of neuroblastoma (NB), and exploring the molecular and cellular mechanisms that underlie neuroblastoma initiation and metastasis. In this review, we will enumerate and illustrate the key advantages of using the zebrafish model in NB research, which allows researchers to: monitor tumor development in real-time; robustly manipulate gene expression (either transiently or stably); rapidly evaluate the cooperative interactions of multiple genetic alterations to disease pathogenesis; and provide a highly efficient and low-cost methodology to screen for effective pharmaceutical interventions (both alone and in combination with one another). This review will then list some of the common challenges of using the zebrafish model and provide strategies for overcoming these difficulties. We have also included visual diagram and figures to illustrate the workflow of cancer model development in zebrafish and provide a summary comparison of commonly used animal models in cancer research, as well as key findings of cooperative contributions between MYCN and diverse singling pathways in NB pathogenesis.


Subject(s)
Neoplasm Proteins/genetics , Nervous System Neoplasms/genetics , Neuroblastoma/genetics , Proto-Oncogene Proteins/genetics , Zebrafish Proteins/genetics , Zebrafish/genetics , Animals , Animals, Genetically Modified , Antineoplastic Agents/pharmacology , Disease Models, Animal , Gene Editing/methods , Gene Expression Regulation, Neoplastic , High-Throughput Screening Assays , Humans , Mutation , Neoplasm Metastasis , Neoplasm Proteins/metabolism , Neoplasm Transplantation/methods , Nervous System Neoplasms/drug therapy , Nervous System Neoplasms/metabolism , Nervous System Neoplasms/pathology , Neuroblastoma/drug therapy , Neuroblastoma/metabolism , Neuroblastoma/pathology , Proto-Oncogene Proteins/metabolism , Signal Transduction , Zebrafish/metabolism , Zebrafish Proteins/metabolism
19.
J Neurosci ; 29(36): 11304-15, 2009 Sep 09.
Article in English | MEDLINE | ID: mdl-19741137

ABSTRACT

Type III neuregulins exposed on axon surfaces control myelination of the peripheral nervous system. It has been shown, for example, that threshold levels of type III beta1a neuregulin dictate not only the myelination fate of axons but also myelin thickness. Here we show that another neuregulin isoform, type III-beta3, plays a distinct role in myelination. Neuronal overexpression of this isoform in mice stimulates Schwann cell proliferation and dramatically enlarges peripheral nerves and ganglia-which come to resemble plexiform neurofibromas-but have no effect on myelin thickness. The nerves display other neurofibroma-like properties, such as abundant collagen fibrils and abundant dissociated Schwann cells that in some cases produce big tumors. Moreover, the organization of Remak bundles is dramatically altered; the small-caliber axons of each bundle are no longer segregated from one another within the cytoplasm of a nonmyelinating Schwann cell but instead are close packed and the whole bundle wrapped as a single unit, frequently by a compact myelin sheath. Because Schwann cell hyperproliferation and Remak bundle degeneration are early hallmarks of type I neurofibromatosis, we suggest that sustained activation of the neuregulin pathway in Remak bundles can contribute to neurofibroma development.


Subject(s)
Axons/physiology , Cell Proliferation , Myelin Sheath/physiology , Neurofibroma/metabolism , Neuroglia/physiology , Schwann Cells/physiology , Animals , Animals, Newborn , Female , Humans , Intracellular Signaling Peptides and Proteins/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nervous System Neoplasms/metabolism , Nervous System Neoplasms/pathology , Neuregulins , Neurofibroma/pathology , Neuroglia/pathology , Pregnancy , Schwann Cells/pathology , Signal Transduction/physiology
20.
Int J Cancer ; 127(4): 805-19, 2010 Aug 15.
Article in English | MEDLINE | ID: mdl-20013810

ABSTRACT

The distance of nerve invasion is an important prognostic factor in pancreatic cancer. The extracellular matrix (ECM) of nerve, mainly composed of laminin, collagen IV and anchoring fibrils, might affect nerve invasion. However, this relationship has not been demonstrated. Our study aimed at discovering the promoting factor of nerve invasion within the tumoral ECM. An animal model was established to evaluate the distance of nerve invasion in murine sciatic nerves by intraneural injection of 6 human pancreatic cancer cell lines. mRNA expression of laminins and anchoring fibrils was compared to the distance of nerve invasion for each cancer cell line. A target molecule provided the strong association between mRNA expression and the distance of nerve invasion. To evaluate the role of a target molecule in nerve invasion, protein expression and function were examined using an animal model and surgical cases. Cancer cells with high laminin gamma2 mRNA and protein expression in their basement membranes were associated with long nerve invasion. Knockdown of laminin gamma2 in cancer cells significantly shortened nerve invasion in the animal model. In 75 patients with pancreatic cancer, a large distance of nerve invasion was associated with high expression levels of laminin gamma2 mRNA and basement membranous deposition of laminin gamma2 protein. Our results indicate that laminin gamma2 plays an important role in nerve invasion. The measurement of the nerve invasion distance in our mouse nerve invasion model is useful for evaluating the molecular mechanisms of nerve invasion.


Subject(s)
Biomarkers, Tumor/genetics , Laminin/genetics , Nervous System Neoplasms/pathology , Pancreatic Neoplasms/pathology , Sciatic Nerve/pathology , Animals , Basement Membrane/metabolism , Biomarkers, Tumor/metabolism , Blotting, Western , Cell Adhesion , Cell Movement , Cell Proliferation , Collagen Type IV/genetics , Collagen Type IV/metabolism , Disease Models, Animal , Female , Gene Expression Profiling , Humans , Immunoenzyme Techniques , Laminin/metabolism , Mice , Mice, SCID , Neoplasm Invasiveness , Nervous System Neoplasms/metabolism , Oligonucleotide Array Sequence Analysis , RNA, Messenger/genetics , RNA, Small Interfering/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL