Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 80
Filter
Add more filters

Publication year range
1.
J Neurooncol ; 168(2): 317-332, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38630384

ABSTRACT

INTRODUCTION: Patients with pediatric low-grade gliomas (pLGGs), the most common primary brain tumors in children, can often benefit from MAPK inhibitor (MAPKi) treatment. However, rapid tumor regrowth, also referred to as rebound growth, may occur once treatment is stopped, constituting a significant clinical challenge. METHODS: Four patient-derived pediatric glioma models were investigated to model rebound growth in vitro based on viable cell counts in response to MAPKi treatment and withdrawal. A multi-omics dataset (RNA sequencing and LC-MS/MS based phospho-/proteomics) was generated to investigate possible rebound-driving mechanisms. Following in vitro validation, putative rebound-driving mechanisms were validated in vivo using the BT-40 orthotopic xenograft model. RESULTS: Of the tested models, only a BRAFV600E-driven model (BT-40, with additional CDKN2A/Bdel) showed rebound growth upon MAPKi withdrawal. Using this model, we identified a rapid reactivation of the MAPK pathway upon MAPKi withdrawal in vitro, also confirmed in vivo. Furthermore, transient overactivation of key MAPK molecules at transcriptional (e.g. FOS) and phosphorylation (e.g. pMEK) levels, was observed in vitro. Additionally, we detected increased expression and secretion of cytokines (CCL2, CX3CL1, CXCL10 and CCL7) upon MAPKi treatment, maintained during early withdrawal. While increased cytokine expression did not have tumor cell intrinsic effects, presence of these cytokines in conditioned media led to increased attraction of microglia cells in vitro. CONCLUSION: Taken together, these data indicate rapid MAPK reactivation upon MAPKi withdrawal as a tumor cell intrinsic rebound-driving mechanism. Furthermore, increased secretion of microglia-recruiting cytokines may play a role in treatment response and rebound growth upon withdrawal, warranting further evaluation.


Subject(s)
Brain Neoplasms , Cytokines , Glioma , Microglia , Mutation , Protein Kinase Inhibitors , Proto-Oncogene Proteins B-raf , Humans , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Microglia/metabolism , Microglia/drug effects , Glioma/metabolism , Glioma/drug therapy , Glioma/pathology , Glioma/genetics , Cytokines/metabolism , Animals , Brain Neoplasms/metabolism , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Brain Neoplasms/genetics , Protein Kinase Inhibitors/pharmacology , Xenograft Model Antitumor Assays , Child , Mice , Cell Proliferation/drug effects , Cell Line, Tumor , MAP Kinase Signaling System/drug effects
2.
Genes Dev ; 30(19): 2187-2198, 2016 Oct 01.
Article in English | MEDLINE | ID: mdl-27737960

ABSTRACT

Oncogene-induced senescence (OIS) is a potent tumor suppressor mechanism. To identify senescence regulators relevant to cancer, we screened an shRNA library targeting genes deleted in hepatocellular carcinoma (HCC). Here, we describe how knockdown of the SWI/SNF component ARID1B prevents OIS and cooperates with RAS to induce liver tumors. ARID1B controls p16INK4a and p21CIP1a transcription but also regulates DNA damage, oxidative stress, and p53 induction, suggesting that SWI/SNF uses additional mechanisms to regulate senescence. To systematically identify SWI/SNF targets regulating senescence, we carried out a focused shRNA screen. We discovered several new senescence regulators, including ENTPD7, an enzyme that hydrolyses nucleotides. ENTPD7 affects oxidative stress, DNA damage, and senescence. Importantly, expression of ENTPD7 or inhibition of nucleotide synthesis in ARID1B-depleted cells results in re-establishment of senescence. Our results identify novel mechanisms by which epigenetic regulators can affect tumor progression and suggest that prosenescence therapies could be employed against SWI/SNF-mutated cancers.


Subject(s)
Carcinoma, Hepatocellular/genetics , Cellular Senescence/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Neoplastic/genetics , Liver Neoplasms/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Animals , Apyrase/metabolism , Carcinoma, Hepatocellular/enzymology , Cell Line , Cell Line, Tumor , Epigenesis, Genetic/genetics , Female , Humans , Liver Neoplasms/enzymology , Male , Mice , Mice, Inbred C57BL , Mutation , RNA, Small Interfering/genetics
3.
Cell Mol Life Sci ; 78(10): 4521-4544, 2021 May.
Article in English | MEDLINE | ID: mdl-34019103

ABSTRACT

Cellular senescence is a process that can prevent tumour development in a cell autonomous manner by imposing a stable cell cycle arrest after oncogene activation. Paradoxically, senescence can also promote tumour growth cell non-autonomously by creating a permissive tumour microenvironment that fuels tumour initiation, progression to malignancy and metastasis. In a pituitary tumour known as adamantinomatous craniopharyngioma (ACP), cells that carry oncogenic ß-catenin mutations and overactivate the WNT signalling pathway form cell clusters that become senescent and activate a senescence-associated secretory phenotype (SASP). Research in mouse models of ACP has provided insights into the function of the senescent cell clusters and revealed a critical role for SASP-mediated activities in paracrine tumour initiation. In this review, we first discuss this research on ACP and subsequently explore the theme of paracrine tumourigenesis in other tumour models available in the literature. Evidence is accumulating supporting the notion that paracrine signalling brought about by senescent cells may underlie tumourigenesis across different tumours and cancer models.


Subject(s)
Carcinogenesis/pathology , Cellular Senescence/physiology , Craniopharyngioma/pathology , Paracrine Communication/physiology , Animals , Humans , Signal Transduction/physiology , Tumor Microenvironment/physiology
4.
Neuropathol Appl Neurobiol ; 47(3): 359-378, 2021 04.
Article in English | MEDLINE | ID: mdl-33378554

ABSTRACT

The study of cell senescence is a burgeoning field. Senescent cells can modify the cellular microenvironment through the secretion of a plethora of biologically active products referred to as the senescence-associated secretory phenotype (SASP). The consequences of these paracrine signals can be either beneficial for tissue homeostasis, if senescent cells are properly cleared and SASP activation is transient, or result in organ dysfunction, when senescent cells accumulate within the tissues and SASP activation is persistent. Several studies have provided evidence for the role of senescence and SASP in promoting age-related diseases or driving organismal ageing. The hype about senescence has been further amplified by the fact that a group of drugs, named senolytics, have been used to successfully ameliorate the burden of age-related diseases and increase health and life span in mice. Ablation of senescent cells in the brain prevents disease progression and improves cognition in murine models of neurodegenerative conditions. The role of senescence in cancer has been more thoroughly investigated, and it is now accepted that senescence is a double-edged sword that can paradoxically prevent or promote tumourigenesis in a context-dependent manner. In addition, senescence induction followed by senolytic treatment is starting to emerge as a novel therapeutic avenue that could improve current anti-cancer therapies and reduce tumour recurrence. In this review, we discuss recent findings supporting the role of cell senescence in the pathogenesis of neurodegenerative diseases and in brain tumours. A better understanding of senescence is likely to result in the development of novel and efficacious anti-senescence therapies against these brain pathologies.


Subject(s)
Brain Neoplasms/pathology , Cellular Senescence/physiology , Nerve Degeneration/pathology , Animals , Carcinogenesis/pathology , Humans
5.
Development ; 144(18): 3289-3302, 2017 09 15.
Article in English | MEDLINE | ID: mdl-28807898

ABSTRACT

Sonic hedgehog (SHH) is an essential morphogenetic signal that dictates cell fate decisions in several developing organs in mammals. In vitro data suggest that SHH is required to specify LHX3+/LHX4+ Rathke's pouch (RP) progenitor identity. However, in vivo studies have failed to reveal such a function, supporting instead a crucial role for SHH in promoting proliferation of these RP progenitors and for differentiation of pituitary cell types. Here, we have used a genetic approach to demonstrate that activation of the SHH pathway is necessary to induce LHX3+/LHX4+ RP identity in mouse embryos. First, we show that conditional deletion of Shh in the anterior hypothalamus results in a fully penetrant phenotype characterised by a complete arrest of RP development, with lack of Lhx3/Lhx4 expression in RP epithelium at 9.0 days post coitum (dpc) and total loss of pituitary tissue by 12.5 dpc. Conversely, overactivation of the SHH pathway by conditional deletion of Ptch1 in RP progenitors leads to severe hyperplasia and enlargement of the Sox2+ stem cell compartment by the end of gestation.


Subject(s)
Cell Lineage , Hedgehog Proteins/metabolism , Hypothalamus/embryology , Hypothalamus/metabolism , LIM-Homeodomain Proteins/metabolism , Pituitary Gland/embryology , Pituitary Gland/metabolism , Transcription Factors/metabolism , Cell Compartmentation , Cell Count , Cell Differentiation , Cell Proliferation , Clone Cells , Crosses, Genetic , Ectoderm/embryology , Ectoderm/metabolism , Embryo, Mammalian/metabolism , Endoderm/embryology , Endoderm/metabolism , Epithelium/embryology , Epithelium/metabolism , Female , Gene Expression Regulation, Developmental , Genotype , Hedgehog Proteins/genetics , Humans , Male , Mutation/genetics , Pituitary Gland/pathology , Signal Transduction , Stem Cells
6.
Development ; 144(12): 2141-2152, 2017 06 15.
Article in English | MEDLINE | ID: mdl-28506993

ABSTRACT

Despite the importance of the RAS-RAF-MAPK pathway in normal physiology and disease of numerous organs, its role during pituitary development and tumourigenesis remains largely unknown. Here, we show that the over-activation of the MAPK pathway, through conditional expression of the gain-of-function alleles BrafV600E and KrasG12D in the developing mouse pituitary, results in severe hyperplasia and abnormal morphogenesis of the gland by the end of gestation. Cell-lineage commitment and terminal differentiation are disrupted, leading to a significant reduction in numbers of most of the hormone-producing cells before birth, with the exception of corticotrophs. Of note, Sox2+ stem cells and clonogenic potential are drastically increased in the mutant pituitaries. Finally, we reveal that papillary craniopharyngioma (PCP), a benign human pituitary tumour harbouring BRAF p.V600E also contains Sox2+ cells with sustained proliferative capacity and disrupted pituitary differentiation. Together, our data demonstrate a crucial function of the MAPK pathway in controlling the balance between proliferation and differentiation of Sox2+ cells and suggest that persistent proliferative capacity of Sox2+ cells may underlie the pathogenesis of PCP.


Subject(s)
Craniopharyngioma/physiopathology , MAP Kinase Signaling System/physiology , Pituitary Neoplasms/physiopathology , Animals , Cell Differentiation , Cell Proliferation , Craniopharyngioma/genetics , Craniopharyngioma/pathology , Embryonic Stem Cells/pathology , Embryonic Stem Cells/physiology , Female , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , MAP Kinase Signaling System/genetics , Mice , Mice, Mutant Strains , Mice, Transgenic , Mutant Proteins/genetics , Mutant Proteins/metabolism , Pituitary Gland/cytology , Pituitary Gland/embryology , Pituitary Gland/enzymology , Pituitary Neoplasms/genetics , Pituitary Neoplasms/pathology , Pregnancy , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , SOXB1 Transcription Factors/metabolism
7.
Neuroendocrinology ; 110(9-10): 797-804, 2020.
Article in English | MEDLINE | ID: mdl-32126562

ABSTRACT

Jakob Erdheim (1874-1937) first described craniopharyn-giomas (CPs) as "hypophyseal duct tumours" and postulated the existence of two tumour types based on their histological features: (1) an aggressive type showing similarities to adamantinomas (tumours of the jaw) and (2) a more benign form characterised by the presence of papillary structures. More than a century later, these initial observations have been confirmed; based on their distinct genetic, epigenetic, and histological features, the WHO classifies CPs into two types: adamantinomatous CPs (ACPs) and papillary CPs (PCPs). Considerable knowledge has been generated on the biology of CPs in the last 20 years. Mutations in CTNNB1 (encoding ß-catenin) are prevalent in ACP, whilst PCPs frequently harbour mutations in BRAF (p.BRAF-V600E). The consequence of these mutations is the activation of either the WNT/ß-catenin (ACP) or the MAPK/ERK (PCP) pathway. Murine models support a critical role for these mutations in tumour formation and have provided important insights into tumour pathogenesis, mostly in ACP. A critical role for cellular senescence has been uncovered in murine models of ACP with relevance to human tumours. Several gene profiling studies of human and murine ACP tumours have identified potential targetable pathways, and novel therapeutic agents are being used in clinical and pre-clinical research, in some cases with excellent results. In this review, we will present the accumulated knowledge on the biological features of these tumours and summarise how these advances are being translated into potential novel treatments.


Subject(s)
Craniopharyngioma , Pituitary Neoplasms , Animals , Craniopharyngioma/drug therapy , Craniopharyngioma/genetics , Craniopharyngioma/metabolism , Craniopharyngioma/pathology , Humans , Pituitary Neoplasms/drug therapy , Pituitary Neoplasms/genetics , Pituitary Neoplasms/metabolism , Pituitary Neoplasms/pathology
8.
EMBO Rep ; 19(2): 269-289, 2018 02.
Article in English | MEDLINE | ID: mdl-29263200

ABSTRACT

WDR11 has been implicated in congenital hypogonadotropic hypogonadism (CHH) and Kallmann syndrome (KS), human developmental genetic disorders defined by delayed puberty and infertility. However, WDR11's role in development is poorly understood. Here, we report that WDR11 modulates the Hedgehog (Hh) signalling pathway and is essential for ciliogenesis. Disruption of WDR11 expression in mouse and zebrafish results in phenotypic characteristics associated with defective Hh signalling, accompanied by dysgenesis of ciliated tissues. Wdr11-null mice also exhibit early-onset obesity. We find that WDR11 shuttles from the cilium to the nucleus in response to Hh signalling. WDR11 regulates the proteolytic processing of GLI3 and cooperates with the transcription factor EMX1 in the induction of downstream Hh pathway gene expression and gonadotrophin-releasing hormone production. The CHH/KS-associated human mutations result in loss of function of WDR11. Treatment with the Hh agonist purmorphamine partially rescues the WDR11 haploinsufficiency phenotypes. Our study reveals a novel class of ciliopathy caused by WDR11 mutations and suggests that CHH/KS may be a part of the human ciliopathy spectrum.


Subject(s)
Ciliopathies/genetics , Ciliopathies/metabolism , Hedgehog Proteins/metabolism , Kallmann Syndrome/genetics , Kallmann Syndrome/metabolism , Membrane Proteins/metabolism , Signal Transduction , Animals , Biopsy , Gene Expression , Gene Expression Profiling , Gene Knockout Techniques , Genetic Association Studies , Genotype , Humans , Kallmann Syndrome/diagnosis , Magnetic Resonance Imaging , Membrane Proteins/genetics , Mice , Mice, Knockout , Mutation , Organ Specificity/genetics , Patched-1 Receptor/genetics , Phenotype , Promoter Regions, Genetic , Protein Binding , Protein Transport , Transcriptome , Zebrafish
9.
Proc Natl Acad Sci U S A ; 113(5): E548-57, 2016 Feb 02.
Article in English | MEDLINE | ID: mdl-26764381

ABSTRACT

Aberrant embryonic development of the hypothalamus and/or pituitary gland in humans results in congenital hypopituitarism (CH). Transcription factor 7-like 1 (TCF7L1), an important regulator of the WNT/ß-catenin signaling pathway, is expressed in the developing forebrain and pituitary gland, but its role during hypothalamo-pituitary (HP) axis formation or involvement in human CH remains elusive. Using a conditional genetic approach in the mouse, we first demonstrate that TCF7L1 is required in the prospective hypothalamus to maintain normal expression of the hypothalamic signals involved in the induction and subsequent expansion of Rathke's pouch progenitors. Next, we reveal that the function of TCF7L1 during HP axis development depends exclusively on the repressing activity of TCF7L1 and does not require its interaction with ß-catenin. Finally, we report the identification of two independent missense variants in human TCF7L1, p.R92P and p.R400Q, in a cohort of patients with forebrain and/or pituitary defects. We demonstrate that these variants exhibit reduced repressing activity in vitro and in vivo relative to wild-type TCF7L1. Together, our data provide support for a conserved molecular function of TCF7L1 as a transcriptional repressor during HP axis development in mammals and identify variants in this transcription factor that are likely to contribute to the etiology of CH.


Subject(s)
Hypothalamo-Hypophyseal System , Transcription Factor 7-Like 1 Protein/physiology , Animals , Cohort Studies , Humans , Mice , Pituitary Gland/abnormalities , Pituitary Gland/metabolism , Pituitary Gland/physiopathology , Prosencephalon/abnormalities , Prosencephalon/metabolism
10.
Br J Cancer ; 118(10): 1283-1288, 2018 05.
Article in English | MEDLINE | ID: mdl-29670296

ABSTRACT

Senescent cells activate genetic programmes that irreversibly inhibit cellular proliferation, but also endow these cells with distinctive metabolic and signalling phenotypes. Although senescence has historically been considered a protective mechanism against tumourigenesis, the activities of senescent cells are increasingly being associated with age-related diseases, including cancer. An important feature of senescent cells is the secretion of a vast array of pro-inflammatory cytokines, chemokines, and growth factors collectively known as the senescence-associated secretory phenotype (SASP). Recent research has shown that SASP paracrine signalling can mediate several pro-tumourigenic effects, such as enhancing malignant phenotypes and promoting tumour initiation. In this review, we summarise the paracrine activities of senescent cells and their role in tumourigenesis through direct effects on growth and proliferation of tumour cells, tumour angiogenesis, invasion and metastasis, cellular reprogramming and emergence of tumour-initiating cells, and tumour interactions with the local immune environment. The evidence described here suggests cellular senescence acts as a double-edged sword in cancer pathogenesis, which demands further attention in order to support the use of senolytic or SASP-modulating compounds for cancer treatment.


Subject(s)
Carcinogenesis/genetics , Cellular Senescence/genetics , Neoplasms/genetics , Paracrine Communication/genetics , Cell Movement/genetics , Cell Proliferation/genetics , Cellular Reprogramming/genetics , Humans , Neoplasm Invasiveness/genetics , Neoplasm Metastasis , Neoplasms/pathology
11.
Acta Neuropathol ; 135(5): 757-777, 2018 05.
Article in English | MEDLINE | ID: mdl-29541918

ABSTRACT

Adamantinomatous craniopharyngiomas (ACPs) are clinically challenging tumours, the majority of which have activating mutations in CTNNB1. They are histologically complex, showing cystic and solid components, the latter comprised of different morphological cell types (e.g. ß-catenin-accumulating cluster cells and palisading epithelium), surrounded by a florid glial reaction with immune cells. Here, we have carried out RNA sequencing on 18 ACP samples and integrated these data with an existing ACP transcriptomic dataset. No studies so far have examined the patterns of gene expression within the different cellular compartments of the tumour. To achieve this goal, we have combined laser capture microdissection with computational analyses to reveal groups of genes that are associated with either epithelial tumour cells (clusters and palisading epithelium), glial tissue or immune infiltrate. We use these human ACP molecular signatures and RNA-Seq data from two ACP mouse models to reveal that cell clusters are molecularly analogous to the enamel knot, a critical signalling centre controlling normal tooth morphogenesis. Supporting this finding, we show that human cluster cells express high levels of several members of the FGF, TGFB and BMP families of secreted factors, which signal to neighbouring cells as evidenced by immunostaining against the phosphorylated proteins pERK1/2, pSMAD3 and pSMAD1/5/9 in both human and mouse ACP. We reveal that inhibiting the MAPK/ERK pathway with trametinib, a clinically approved MEK inhibitor, results in reduced proliferation and increased apoptosis in explant cultures of human and mouse ACP. Finally, we analyse a prominent molecular signature in the glial reactive tissue to characterise the inflammatory microenvironment and uncover the activation of inflammasomes in human ACP. We validate these results by immunostaining against immune cell markers, cytokine ELISA and proteome analysis in both solid tumour and cystic fluid from ACP patients. Our data support a new molecular paradigm for understanding ACP tumorigenesis as an aberrant mimic of natural tooth development and opens new therapeutic opportunities by revealing the activation of the MAPK/ERK and inflammasome pathways in human ACP.


Subject(s)
Craniopharyngioma/metabolism , MAP Kinase Signaling System , Pituitary Neoplasms/metabolism , Transcriptome , Tumor Microenvironment/physiology , Animals , Computational Biology , Craniopharyngioma/pathology , Craniopharyngioma/therapy , Cytokines/metabolism , Disease Models, Animal , Humans , Inflammation/metabolism , Inflammation/therapy , Laser Capture Microdissection , Mice , Neuroglia/metabolism , Odontogenesis/physiology , Pituitary Gland/embryology , Pituitary Gland/pathology , Pituitary Neoplasms/pathology , Pituitary Neoplasms/therapy , Sequence Analysis, RNA , Tissue Culture Techniques
12.
Stem Cells ; 34(2): 268-76, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26763580

ABSTRACT

The existence of tissue-specific progenitor/stem cells in the adult pituitary gland of the mouse has been demonstrated recently using genetic tracing experiments. These cells have the capacity to differentiate into all of the different cell lineages of the anterior pituitary and self-propagate in vitro and can therefore contribute to normal homeostasis of the gland. In addition, they play a critical role in tumor formation, specifically in the etiology of human adamantinomatous craniopharyngioma, a clinically relevant tumor that is associated with mutations in CTNNB1 (gene encoding ß-catenin). Mouse studies have shown that only pituitary embryonic precursors or adult stem cells are able to generate tumors when targeted with oncogenic ß-catenin, suggesting that the cell context is critical for mutant ß-catenin to exert its oncogenic effect. Surprisingly, the bulk of the tumor cells are not derived from the mutant progenitor/stem cells, suggesting that tumors are induced in a paracrine manner. Therefore, the cell sustaining the mutation in ß-catenin and the cell-of-origin of the tumors are different. In this review, we will discuss the in vitro and in vivo evidence demonstrating the presence of stem cells in the adult pituitary and analyze the evidence showing a potential role of these stem cells in pituitary tumors.


Subject(s)
Craniopharyngioma/metabolism , Mutation , Neoplasm Proteins/metabolism , Neoplastic Stem Cells/metabolism , Paracrine Communication , Pituitary Neoplasms/metabolism , beta Catenin/metabolism , Animals , Craniopharyngioma/genetics , Craniopharyngioma/pathology , Humans , Mice , Neoplasm Proteins/genetics , Neoplastic Stem Cells/pathology , Pituitary Neoplasms/pathology , beta Catenin/genetics
13.
Neurosurg Focus ; 41(6): E4, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27903120

ABSTRACT

Since the first identification of CTNNB1 mutations in adamantinomatous craniopharyngioma (ACP), much has been learned about the molecular pathways and processes that are disrupted in ACP pathogenesis. To date this understanding has not translated into tangible patient benefit. The recent development of novel techniques and a range of preclinical models now provides an opportunity to begin to support treatment decisions and develop new therapeutics based on molecular pathology. In this review the authors summarize many of the key findings and pathways implicated in ACP pathogenesis and discuss the challenges that need to be tackled to translate these basic science findings for the benefit of patients.


Subject(s)
Biomarkers, Tumor/genetics , Craniopharyngioma/genetics , Craniopharyngioma/pathology , Pituitary Neoplasms/genetics , Pituitary Neoplasms/pathology , Humans , beta Catenin/genetics
14.
Neuropathol Appl Neurobiol ; 41(6): 721-32, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25611703

ABSTRACT

Adamantinomatous craniopharyngiomas (ACPs) are the most common pituitary tumours in children. Although histologically benign, these are clinically aggressive tumours, difficult to manage and associated with poor quality of life for the patients. Several human and mouse studies have provided unequivocal evidence that the over-activation of the WNT/ß-catenin signalling pathway underlies the molecular aetiology of these tumours. Recently, research using genetically modified mouse models of human ACP have revealed a critical and unexpected non-cell autonomous role for pituitary stem cells in ACP tumourigenesis, which has expanded the cancer stem cell paradigm. As the result of this basic research, the pathogenesis of ACP is being unveiled, with promising implications for the development of novel treatments against these childhood neoplasms. These benign tumours may additionally represent a unique model to provide insights into the initial steps of oncogenesis.


Subject(s)
Craniopharyngioma/genetics , Craniopharyngioma/metabolism , Pituitary Neoplasms/genetics , Pituitary Neoplasms/metabolism , Animals , Craniopharyngioma/pathology , Humans , Mice , Neoplastic Stem Cells/metabolism , Pituitary Neoplasms/pathology , Stem Cells/metabolism , Wnt Signaling Pathway/genetics , beta Catenin/metabolism
15.
Development ; 138(22): 4931-42, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22007134

ABSTRACT

The Wnt/ß-catenin pathway plays an essential role during regionalisation of the vertebrate neural plate and its inhibition in the most anterior neural ectoderm is required for normal forebrain development. Hesx1 is a conserved vertebrate-specific transcription factor that is required for forebrain development in Xenopus, mice and humans. Mouse embryos deficient for Hesx1 exhibit a variable degree of forebrain defects, but the molecular mechanisms underlying these defects are not fully understood. Here, we show that injection of a hesx1 morpholino into a 'sensitised' zygotic headless (tcf3) mutant background leads to severe forebrain and eye defects, suggesting an interaction between Hesx1 and the Wnt pathway during zebrafish forebrain development. Consistent with a requirement for Wnt signalling repression, we highlight a synergistic gene dosage-dependent interaction between Hesx1 and Tcf3, a transcriptional repressor of Wnt target genes, to maintain anterior forebrain identity during mouse embryogenesis. In addition, we reveal that Tcf3 is essential within the neural ectoderm to maintain anterior character and that its interaction with Hesx1 ensures the repression of Wnt targets in the developing forebrain. By employing a conditional loss-of-function approach in mouse, we demonstrate that deletion of ß-catenin, and concomitant reduction of Wnt signalling in the developing anterior forebrain of Hesx1-deficient embryos, leads to a significant rescue of the forebrain defects. Finally, transcriptional profiling of anterior forebrain precursors from mouse embryos expressing eGFP from the Hesx1 locus provides molecular evidence supporting a novel function of Hesx1 in mediating repression of Wnt/ß-catenin target activation in the developing forebrain.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/physiology , Gene Expression Regulation, Developmental , Homeodomain Proteins/physiology , Prosencephalon/embryology , Repressor Proteins/physiology , Wnt Signaling Pathway/genetics , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cells, Cultured , Down-Regulation/genetics , Embryo, Mammalian , Female , Gene Expression Profiling , Gene Expression Regulation, Developmental/physiology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microarray Analysis , Prosencephalon/metabolism , Prosencephalon/physiology , Repressor Proteins/genetics , Repressor Proteins/metabolism , Wnt Signaling Pathway/physiology , beta Catenin/genetics , beta Catenin/physiology
16.
Brain ; 136(Pt 10): 3096-105, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24022475

ABSTRACT

We describe a previously unreported syndrome characterized by secondary (post-natal) microcephaly with fronto-temporal lobe hypoplasia, multiple pituitary hormone deficiency, seizures, severe visual impairment and abnormalities of the kidneys and urinary tract in a highly consanguineous family with six affected children. Homozygosity mapping and exome sequencing revealed a novel homozygous frameshift mutation in the basic helix-loop-helix transcription factor gene ARNT2 (c.1373_1374dupTC) in affected individuals. This mutation results in absence of detectable levels of ARNT2 transcript and protein from patient fibroblasts compared with controls, consistent with nonsense-mediated decay of the mutant transcript and loss of ARNT2 function. We also show expression of ARNT2 within the central nervous system, including the hypothalamus, as well as the renal tract during human embryonic development. The progressive neurological abnormalities, congenital hypopituitarism and post-retinal visual pathway dysfunction in affected individuals demonstrates for the first time the essential role of ARNT2 in the development of the hypothalamo-pituitary axis, post-natal brain growth, and visual and renal function in humans.


Subject(s)
Aryl Hydrocarbon Receptor Nuclear Translocator/genetics , Basic Helix-Loop-Helix Transcription Factors/genetics , Hypopituitarism/genetics , Kidney/abnormalities , Microcephaly/genetics , Mutation/genetics , Pituitary Hormones/metabolism , Visual Perception , Child , Child, Preschool , Female , Humans , Hypopituitarism/diagnosis , Hypothalamus/metabolism , Kidney/metabolism , Male , Microcephaly/diagnosis , Pituitary Hormones/genetics , Syndrome , Transcription Factors
17.
Cell Mol Life Sci ; 70(20): 3739-52, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23397132

ABSTRACT

Research from the last 15 years has provided a working model for how the anterior forebrain is induced and specified during the early stages of embryogenesis. This model relies on three basic processes: (1) induction of the neural plate from naive ectoderm requires the inhibition of BMP/TGFß signaling; (2) induced neural tissue initially acquires an anterior identity (i.e., anterior forebrain); (3) maintenance and expansion of the anterior forebrain depends on the antagonism of posteriorizing signals that would otherwise transform this tissue into posterior neural fates. In this review, we present a historical perspective examining some of the significant experiments that have helped to delineate this molecular model. In addition, we discuss the function of the relevant tissues that act prior to and during gastrulation to ensure proper anterior forebrain formation. Finally, we elaborate data, mainly obtained from the analyses of mouse mutants, supporting a role for transcriptional repressors in the regulation of cell competence within the anterior forebrain. The aim of this review is to provide the reader with a general overview of the signals as well as the signaling centers that control the development of the anterior neural plate.


Subject(s)
Body Patterning , Gastrulation , Gene Expression Regulation, Developmental , Prosencephalon/metabolism , Animals , Endoderm/embryology , Endoderm/metabolism , Gastrula/embryology , Gastrula/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Mice , Models, Molecular , Neural Plate/embryology , Neural Plate/metabolism , Prosencephalon/embryology , Repressor Proteins/genetics , Repressor Proteins/metabolism , Wnt Signaling Pathway
18.
Proc Natl Acad Sci U S A ; 108(28): 11482-7, 2011 Jul 12.
Article in English | MEDLINE | ID: mdl-21636786

ABSTRACT

Wingless (Wnt)/ß-catenin signaling plays an essential role during normal development, is a critical regulator of stem cells, and has been associated with cancer in many tissues. Here we demonstrate that genetic expression of a degradation-resistant mutant form of ß-catenin in early Rathke's pouch (RP) progenitors leads to pituitary hyperplasia and severe disruption of the pituitary-specific transcription factor 1-lineage differentiation resulting in extreme growth retardation and hypopituitarism. Mutant mice mostly die perinatally, but those that survive weaning develop lethal pituitary tumors, which closely resemble human adamantinomatous craniopharyngioma, an epithelial tumor associated with mutations in the human ß-catenin gene. The tumorigenic effect of mutant ß-catenin is observed only when expressed in undifferentiated RP progenitors, but tumors do not form when committed or differentiated cells are targeted to express this protein. Analysis of affected pituitaries indicates that expression of mutant ß-catenin leads to a significant increase in the total numbers of pituitary progenitor/stem cells as well as in their proliferation potential. Our findings provide insights into the role of the Wnt pathway in normal pituitary development and demonstrate a causative role for mutated ß-catenin in an undifferentiated RP progenitor in the genesis of murine and human craniopharyngioma.


Subject(s)
Pituitary Gland/cytology , Pituitary Gland/metabolism , Pituitary Neoplasms/etiology , Pituitary Neoplasms/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Wnt Proteins/metabolism , Animals , Cell Differentiation , Craniopharyngioma/etiology , Craniopharyngioma/genetics , Craniopharyngioma/metabolism , Craniopharyngioma/pathology , Disease Models, Animal , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Mice , Mice, Mutant Strains , Mutant Proteins/genetics , Mutant Proteins/metabolism , Pituitary Gland/growth & development , Pituitary Neoplasms/genetics , Pituitary Neoplasms/pathology , Repressor Proteins/genetics , Repressor Proteins/metabolism , Signal Transduction , beta Catenin/genetics , beta Catenin/metabolism
19.
EBioMedicine ; 99: 104905, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38043401

ABSTRACT

The long-standing view of senescent cells as passive and dysfunctional biological remnants has recently shifted into a new paradigm where they are main players in the development of many diseases, including cancer. The senescence programme represents a first line of defence that prevents tumour cell growth but also leads to the secretion of multiple pro-inflammatory and pro-tumourigenic factors that fuel tumour initiation, growth, and progression. Here, we review the main molecular features and biological functions of senescent cells in cancer, including the outcomes of inducing or targeting senescence. We discuss evidence on the role of cellular senescence in pituitary tumours, with an emphasis on adamantinomatous craniopharyngioma (ACP) and pituitary adenomas. Although senescence has been proposed to be a tumour-preventing mechanism in pituitary adenomas, research in ACP has shown that senescent cells are tumour-promoting in both murine models and human tumours. Future studies characterizing the impact of targeting senescent cells may result in novel therapies against pituitary tumours.


Subject(s)
Craniopharyngioma , Pituitary Neoplasms , Humans , Mice , Child , Animals , Pituitary Neoplasms/genetics , Pituitary Gland , Craniopharyngioma/genetics , Craniopharyngioma/pathology , Carcinogenesis/pathology , Cell Transformation, Neoplastic/pathology , Cellular Senescence
20.
Neuro Oncol ; 26(6): 1109-1123, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38334125

ABSTRACT

BACKGROUND: Cellular senescence can have positive and negative effects on the body, including aiding in damage repair and facilitating tumor growth. Adamantinomatous craniopharyngioma (ACP), the most common pediatric sellar/suprasellar brain tumor, poses significant treatment challenges. Recent studies suggest that senescent cells in ACP tumors may contribute to tumor growth and invasion by releasing a senesecence-associated secretory phenotype. However, a detailed analysis of these characteristics has yet to be completed. METHODS: We analyzed primary tissue samples from ACP patients using single-cell, single-nuclei, and spatial RNA sequencing. We performed various analyses, including gene expression clustering, inferred senescence cells from gene expression, and conducted cytokine signaling inference. We utilized LASSO to select essential gene expression pathways associated with senescence. Finally, we validated our findings through immunostaining. RESULTS: We observed significant diversity in gene expression and tissue structure. Key factors such as NFKB, RELA, and SP1 are essential in regulating gene expression, while senescence markers are present throughout the tissue. SPP1 is the most significant cytokine signaling network among ACP cells, while the Wnt signaling pathway predominantly occurs between epithelial and glial cells. Our research has identified links between senescence-associated features and pathways, such as PI3K/Akt/mTOR, MYC, FZD, and Hedgehog, with increased P53 expression associated with senescence in these cells. CONCLUSIONS: A complex interplay between cellular senescence, cytokine signaling, and gene expression pathways underlies ACP development. Further research is crucial to understand how these elements interact to create novel therapeutic approaches for patients with ACP.


Subject(s)
Cellular Senescence , Craniopharyngioma , Machine Learning , Pituitary Neoplasms , Humans , Craniopharyngioma/metabolism , Craniopharyngioma/pathology , Craniopharyngioma/genetics , Pituitary Neoplasms/pathology , Pituitary Neoplasms/metabolism , Pituitary Neoplasms/genetics , Biomarkers, Tumor/metabolism , Biomarkers, Tumor/genetics , Phenotype , Gene Expression Regulation, Neoplastic , Child , Male , Female
SELECTION OF CITATIONS
SEARCH DETAIL