Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
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
2.
Ground Water ; 62(2): 174-183, 2024.
Article in English | MEDLINE | ID: mdl-37482948

ABSTRACT

Chemical and isotopic processes occur in every segment of the hydrological cycle. Hydrogeochemistry-the subdiscipline that studies these processes-has seen a transformation from "witch's brew" to credible science since 2000. Going forward, hydrogeochemical research and applications are critical to meeting urgent societal needs of climate change mitigation and clean energy, such as (1) removing CO2 from the atmosphere and storing gigatons of CO2 in soils and aquifers to achieve net-zero emissions, (2) securing critical minerals in support of the transition from fossil fuels to renewable energies, and (3) protecting water resources by adapting to a warming climate. In the last two decades, we have seen extensive activity and progress in four research areas of hydrogeochemistry related to water-rock interactions: arsenic contamination of groundwater; the use of isotopic and chemical tracers to quantify groundwater recharge and submarine groundwater discharge; the kinetics of chemical reactions and the mineral-water interface's control of contaminant fate and transport; and the transformation of geochemical modeling from an expert-only exercise to a widely accessible tool. In the future, embracing technological advances in machine learning, cyberinfrastructure, and isotope analytical tools will allow breakthrough research and expand the role of hydrogeochemistry in meeting society's needs for climate change mitigation and the transition from fossil fuels to renewable energies.


Subject(s)
Groundwater , Water Pollutants, Chemical , Groundwater/chemistry , Environmental Monitoring , Carbon Dioxide , Water , Fossil Fuels , Water Pollutants, Chemical/analysis
4.
Endocr Rev ; 44(3): 518-538, 2023 05 08.
Article in English | MEDLINE | ID: mdl-36574377

ABSTRACT

Craniopharyngiomas (CPs) are clinically aggressive tumors because of their invasive behavior and recalcitrant tendency to recur after therapy. There are 2 types based on their distinct histology and molecular features: the papillary craniopharyngioma (PCP), which is associated with BRAF-V600E mutations and the adamantinomatous craniopharyngioma (ACP), characterized by mutations in CTNNB1 (encoding ß-catenin). Patients with craniopharyngioma show symptoms linked to the location of the tumor close to the optic pathways, hypothalamus, and pituitary gland, such as increased intracranial pressure, endocrine deficiencies, and visual defects. Treatment is not specific and mostly noncurative, and frequently includes surgery, which may achieve gross total or partial resection, followed by radiotherapy. In cystic tumors, frequent drainage is often required and intracystic instillation of drugs has been used to help manage cyst refilling. More recently targeted therapies have been used, particularly in PCP, but also now in ACP and clinical trials are underway or in development. Although patient survival is high, the consequences of the tumor and its treatment can lead to severe comorbidities resulting in poor quality of life, in particular for those patients who bear tumors with hypothalamic involvement. Accordingly, in these patients at risk for the development of a hypothalamic syndrome, hypothalamus-sparing treatment strategies such as limited resection followed by irradiation are recommended. In this review, we provide an update on various aspects of CP, with emphasis on recent advances in the understanding of tumor pathogenesis, clinical consequences, management, and therapies.


Subject(s)
Craniopharyngioma , Pituitary Neoplasms , Humans , Craniopharyngioma/diagnosis , Craniopharyngioma/genetics , Craniopharyngioma/therapy , Quality of Life , Pituitary Neoplasms/diagnosis , Pituitary Neoplasms/genetics , Pituitary Neoplasms/therapy , Mutation
5.
BMJ Open ; 12(9): e067123, 2022 09 08.
Article in English | MEDLINE | ID: mdl-36378622

ABSTRACT

INTRODUCTION: Gliomas are the most common primary tumour of the central nervous system (CNS), with an estimated annual incidence of 6.6 per 100 000 individuals in the USA and around 14 deaths per day from brain tumours in the UK. The genomic and biological landscape of brain tumours has been increasingly defined and, since 2016, the WHO classification of tumours of the CNS incorporates molecular data, along with morphology, to define tumour subtypes more accurately. The Tessa Jowell BRAIN MATRIX Platform (TJBM) study aims to create a transformative clinical research infrastructure that leverages UK National Health Service resources to support research that is patient centric and attractive to both academic and commercial investors. METHODS AND ANALYSIS: The TJBM study is a programme of work with the principal purpose to improve the knowledge of glioma and treatment for patients with glioma. The programme includes a platform study and subsequent interventional clinical trials (as separate protocols). The platform study described here is the backbone data-repository of disease, treatment and outcome data from clinical, imaging and pathology data being collected in patients with glioma from secondary care hospitals. The primary outcome measure of the platform is time from biopsy to integrated histological-molecular diagnosis using whole-genome sequencing and epigenomic classification. Secondary outcome measures include those that are process centred, patient centred and framework based. Target recruitment for the study is 1000 patients with interim analyses at 100 and 500 patients. ETHICS AND DISSEMINATION: The study will be performed in accordance with the recommendations guiding physicians in biomedical research involving human subjects, adopted by the 18th World Medical Association General Assembly, Helsinki, Finland and stated in the respective participating countries' laws governing human research, and Good Clinical Practice. The protocol was initially approved on 18 February 2020 by West Midlands - Edgbaston Research Ethics Committee; the current protocol (v3.0) was approved on 15 June 2022. Participants will be required to provide written informed consent. A meeting will be held after the end of the study to allow discussion of the main results among the collaborators prior to publication. The results of this study will be disseminated through national and international presentations and peer-reviewed publications. Manuscripts will be prepared by the Study Management Group and authorship will be determined by mutual agreement. TRIAL REGISTRATION NUMBER: NCT04274283, 18-Feb-2020; ISRCTN14218060, 03-Feb-2020.


Subject(s)
Brain Neoplasms , Glioma , Humans , State Medicine , Informed Consent , Glioma/genetics , Glioma/therapy , Brain Neoplasms/genetics , Brain Neoplasms/therapy , Finland
6.
Neurooncol Adv ; 4(1): vdac053, 2022.
Article in English | MEDLINE | ID: mdl-35591977

ABSTRACT

Background: Relapsed ependymoma has a dismal prognosis, and the role of chemotherapy at relapse remains unclear. This study prospectively evaluated the efficacy of intensive intravenous (IV) etoposide in patients less than 21 years of age with relapsed intracranial ependymoma (NCT00278252). Methods: This was a single-arm, open-label, phase II trial using Gehan's two-stage design. Patients received IV etoposide 100 mg/m2 on days 1-3, 8-10, and 15-17 of each 28-day cycle, up to maximum of 6 cycles. Primary outcome was radiological response after 3 cycles. Pharmacokinetic analysis was performed in 10 patients. Results: Twenty-five patients were enrolled and included in the intention-to-treat (ITT) analysis. Three patients were excluded in per-protocol (PP) analysis. After 3 cycles of etoposide, 5 patients (ITT 20%/PP 23%) had a complete response (CR), partial response (PR), or objective response (OR). Nine patients (ITT 36%/PP 41%,) had a best overall response of CR, PR, or OR. 1-year PFS was 24% in ITT and 23% in PP populations. 1-year OS was 56% and 59%, 5-year OS was 20% and 18%, respectively, in ITT and PP populations. Toxicity was predominantly hematological, with 20/25 patients experiencing a grade 3 or higher hematological adverse event. Conclusions: This study confirms the activity of IV etoposide against relapsed ependymoma, however, this is modest, not sustained, and similar to that with oral etoposide, albeit with increased toxicity. These results confirm the dismal prognosis of this disease, provide a rationale to include etoposide within drug combinations, and highlight the need to develop novel treatments for recurrent ependymoma.

7.
Arch Dis Child Educ Pract Ed ; 106(5): 314-316, 2021 10.
Article in English | MEDLINE | ID: mdl-32321737

Subject(s)
Education, Medical , Humans
8.
Stem Cell Reports ; 13(6): 970-979, 2019 12 10.
Article in English | MEDLINE | ID: mdl-31761678

ABSTRACT

The role of the homeobox transcriptional repressor HESX1 in embryonic stem cells (ESCs) remains mostly unknown. Here, we show that Hesx1 is expressed in the preimplantation mouse embryo, where it is required during developmental diapause. Absence of Hesx1 leads to reduced expression of epiblast and primitive endoderm determinants and failure of diapaused embryos to resume embryonic development after implantation. Genetic deletion of Hesx1 impairs self-renewal and promotes differentiation toward epiblast by reducing the expression of pluripotency factors and decreasing the activity of LIF/STAT3 signaling. We reveal that Hesx1-deficient ESCs show elevated ERK pathway activation, resulting in accelerated differentiation toward primitive endoderm, which can be prevented by overexpression of Hesx1. Together, our data provide evidence for a novel role of Hesx1 in the control of self-renewal and maintenance of the undifferentiated state in ESCs and mouse embryos.


Subject(s)
Cell Differentiation/genetics , Cell Self Renewal/genetics , Diapause/genetics , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Gene Deletion , Repressor Proteins/deficiency , Animals , Biomarkers , Embryonic Development , Fluorescent Antibody Technique , Gene Expression Regulation , Homeodomain Proteins , Leukemia Inhibitory Factor/metabolism , MAP Kinase Signaling System , Mice , Models, Biological , Phenotype , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Signal Transduction
9.
F1000Res ; 8: 1544, 2019.
Article in English | MEDLINE | ID: mdl-32047602

ABSTRACT

This manuscript describes the study of two cases of craniopharyngioma, which have been examined repeatedly over three separate centuries. This includes analysis by Josef Engel in 1839, who sought to uncover the physiological role of the pituitary gland; Jacob Erdheim in 1904, who initially described the disease we now call craniopharyngioma, and recent high resolution MRI and micro-CT imaging and attempted DNA analyses of the tumours. The cases highlight how, rightly or wrongly, our interpretation of data is shaped by the technologies, methodologies and prevailing theories of a given time.


Subject(s)
Craniopharyngioma , Pituitary Neoplasms , Craniopharyngioma/diagnosis , Craniopharyngioma/history , History, 19th Century , History, 20th Century , History, 21st Century , Humans , Magnetic Resonance Imaging , Pituitary Neoplasms/diagnosis , Pituitary Neoplasms/history
10.
Endocr Relat Cancer ; 26(1): 215-225, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30139767

ABSTRACT

Tumours of the anterior pituitary can manifest from all endocrine cell types but the mechanisms for determining their specification are not known. The Hippo kinase cascade is a crucial signalling pathway regulating growth and cell fate in numerous organs. There is mounting evidence implicating this in tumour formation, where it is emerging as an anti-cancer target. We previously demonstrated activity of the Hippo kinase cascade in the mouse pituitary and nuclear association of its effectors YAP/TAZ with SOX2-expressing pituitary stem cells. Here, we sought to investigate whether these components are expressed in the human pituitary and if they are deregulated in human pituitary tumours. Analysis of pathway components by immunofluorescence reveals pathway activity during normal human pituitary development and in the adult gland. Poorly differentiated pituitary tumours (null-cell adenomas, adamantinomatous craniopharyngiomas (ACPs) and papillary craniopharyngiomas (PCPs)), displayed enhanced expression of pathway effectors YAP/TAZ. In contrast, differentiated adenomas displayed lower or absent levels. Knockdown of the kinase-encoding Lats1 in GH3 rat mammosomatotropinoma cells suppressed Prl and Gh promoter activity following an increase in YAP/TAZ levels. In conclusion, we have demonstrated activity of the Hippo kinase cascade in the human pituitary and association of high YAP/TAZ with repression of the differentiated state both in vitro and in vivo. Characterisation of this pathway in pituitary tumours is of potential prognostic value, opening up putative avenues for treatments.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Apoptosis Regulatory Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Pituitary Gland/metabolism , Pituitary Neoplasms/metabolism , Trans-Activators/metabolism , Transcription Factors/metabolism , Aged , Aged, 80 and over , Animals , Cell Line, Tumor , Female , Humans , Male , Pituitary Neoplasms/genetics , Protein Serine-Threonine Kinases/genetics , RNA Interference , Rats , Transcriptional Coactivator with PDZ-Binding Motif Proteins , YAP-Signaling Proteins
11.
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
12.
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
13.
Brain Pathol ; 28(4): 475-483, 2018 07.
Article in English | MEDLINE | ID: mdl-28481062

ABSTRACT

To assess the clinical relevance of transgenic and patient-derived xenograft models of adamantinomatous craniopharyngioma (ACP) using serial magnetic resonance imaging (MRI) and high resolution post-mortem microcomputed tomography (µ-CT), with correlation with histology and human ACP imaging. The growth patterns and radiological features of tumors arising in Hesx1Cre/+ ;Ctnnb1lox(ex3)/+ transgenic mice, and of patient-derived ACP xenografts implanted in the cerebral cortex, were monitored longitudinally in vivo with anatomical and functional MRI, and by ex vivo µ-CT at study end. Pathological correlates with hematoxylin and eosin stained sections were investigated. Early enlargement and heterogeneity of Hesx1Cre/+ ;Ctnnb1lox(ex3)/+ mouse pituitaries was evident at initial imaging at 8 weeks, which was followed by enlargement of a solid tumor, and development of cysts and hemorrhage. Tumors demonstrated MRI features that recapitulated those of human ACP, specifically, T1 -weighted signal enhancement in the solid tumor component following Gd-DTPA administration, and in some animals, hyperintense cysts on FLAIR and T1 -weighted images. Ex vivo µ-CT correlated with MRI findings and identified smaller cysts, which were confirmed by histology. Characteristic histological features, including wet keratin and calcification, were visible on µ-CT and verified by histological sections of patient-derived ACP xenografts. The Hesx1Cre/+ ;Ctnnb1lox(ex3)/+ transgenic mouse model and cerebral patient-derived ACP xenografts recapitulate a number of the key radiological features of the human disease and provide promising foundations for in vivo trials of novel therapeutics for the treatment of these tumors.


Subject(s)
Craniopharyngioma/diagnostic imaging , Craniopharyngioma/pathology , Disease Models, Animal , Animals , Craniopharyngioma/genetics , Heterografts/diagnostic imaging , Heterografts/pathology , Homeodomain Proteins/genetics , Humans , Magnetic Resonance Imaging , Male , Mice, Transgenic , Middle Aged , Repressor Proteins/genetics , X-Ray Microtomography , beta Catenin/genetics
14.
Acta Neuropathol ; 135(1): 115-129, 2018 01.
Article in English | MEDLINE | ID: mdl-29058119

ABSTRACT

Glioneuronal tumours are an important cause of treatment-resistant epilepsy. Subtypes of tumour are often poorly discriminated by histological features and may be difficult to diagnose due to a lack of robust diagnostic tools. This is illustrated by marked variability in the reported frequencies across different epilepsy surgical series. To address this, we used DNA methylation arrays and RNA sequencing to assay the methylation and expression profiles within a large cohort of glioneuronal tumours. By adopting a class discovery approach, we were able to identify two distinct groups of glioneuronal tumour, which only partially corresponded to the existing histological classification. Furthermore, by additional molecular analyses, we were able to identify pathogenic mutations in BRAF and FGFR1, specific to each group, in a high proportion of cases. Finally, by interrogating our expression data, we were able to show that each molecular group possessed expression phenotypes suggesting different cellular differentiation: astrocytic in one group and oligodendroglial in the second. Informed by this, we were able to identify CCND1, CSPG4, and PDGFRA as immunohistochemical targets which could distinguish between molecular groups. Our data suggest that the current histological classification of glioneuronal tumours does not adequately represent their underlying biology. Instead, we show that there are two molecular groups within glioneuronal tumours. The first of these displays astrocytic differentiation and is driven by BRAF mutations, while the second displays oligodendroglial differentiation and is driven by FGFR1 mutations.


Subject(s)
Brain Neoplasms/metabolism , Epilepsy/metabolism , Ganglioglioma/metabolism , Neoplasms, Neuroepithelial/metabolism , Adolescent , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Brain Neoplasms/surgery , Child , Child, Preschool , Cohort Studies , DNA Methylation , Epilepsy/genetics , Epilepsy/pathology , Epilepsy/surgery , Female , Ganglioglioma/genetics , Ganglioglioma/pathology , Ganglioglioma/surgery , Gene Expression , Humans , Infant , Male , Mutation , Neoplasms, Neuroepithelial/genetics , Neoplasms, Neuroepithelial/pathology , Neoplasms, Neuroepithelial/surgery , Phenotype , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Receptor, Fibroblast Growth Factor, Type 1/metabolism
15.
Nat Commun ; 8(1): 1819, 2017 11 28.
Article in English | MEDLINE | ID: mdl-29180744

ABSTRACT

Senescent cells may promote tumour progression through the activation of a senescence-associated secretory phenotype (SASP), whether these cells are capable of initiating tumourigenesis in vivo is not known. Expression of oncogenic ß-catenin in Sox2+ young adult pituitary stem cells leads to formation of clusters of stem cells and induction of tumours resembling human adamantinomatous craniopharyngioma (ACP), derived from Sox2- cells in a paracrine manner. Here, we uncover the mechanisms underlying this paracrine tumourigenesis. We show that expression of oncogenic ß-catenin in Hesx1+ embryonic precursors also results in stem cell clusters and paracrine tumours. We reveal that human and mouse clusters are analogous and share a common signature of senescence and SASP. Finally, we show that mice with reduced senescence and SASP responses exhibit decreased tumour-inducing potential. Together, we provide evidence that senescence and a stem cell-associated SASP drive cell transformation and tumour initiation in vivo in an age-dependent fashion.


Subject(s)
Cellular Senescence/physiology , Craniopharyngioma/metabolism , Neoplastic Stem Cells/metabolism , Pituitary Neoplasms/metabolism , Aniline Compounds/pharmacology , Animals , Biphenyl Compounds/pharmacology , Cell Transformation, Neoplastic , Child , Craniopharyngioma/pathology , Disease Models, Animal , Homeodomain Proteins/metabolism , Humans , Mice , Nitrophenols/pharmacology , Oncogenes/physiology , Piperazines/pharmacology , Pituitary Gland/metabolism , Pituitary Gland/pathology , Pituitary Neoplasms/pathology , Repressor Proteins/metabolism , SOXB1 Transcription Factors/metabolism , Sulfonamides/pharmacology , Exome Sequencing , Young Adult , beta Catenin/metabolism
16.
J Neuropathol Exp Neurol ; 76(9): 779-788, 2017 Sep 01.
Article in English | MEDLINE | ID: mdl-28859336

ABSTRACT

Pediatric adamantinomatous craniopharyngioma (ACP) is a highly solid and cystic tumor, often causing substantial damage to critical neuroendocrine structures such as the hypothalamus, pituitary gland, and optic apparatus. Paracrine signaling mechanisms driving tumor behavior have been hypothesized, with IL-6R overexpression identified as a potential therapeutic target. To identify potential novel therapies, we characterized inflammatory and immunomodulatory factors in ACP cyst fluid and solid tumor components. Cytometric bead analysis revealed a highly pro-inflammatory cytokine pattern in fluid from ACP compared to fluids from another cystic pediatric brain tumor, pilocytic astrocytoma. Cytokines and chemokines with particularly elevated concentrations in ACPs were IL-6, CXCL1 (GRO), CXCL8 (IL-8) and the immunosuppressive cytokine IL-10. These data were concordant with solid tumor compartment transcriptomic data from a larger cohort of ACPs, other pediatric brain tumors and normal brain. The majority of receptors for these cytokines and chemokines were also over-expressed in ACPs. In addition to IL-10, the established immunosuppressive factor IDO-1 was overexpressed by ACPs at the mRNA and protein levels. These data indicate that ACP cyst fluids and solid tumor components are characterized by an inflammatory cytokine and chemokine expression pattern. Further study regarding selective cytokine blockade may inform novel therapeutic interventions.


Subject(s)
Craniopharyngioma/metabolism , Cyst Fluid/metabolism , Cytokines/metabolism , Pituitary Neoplasms/metabolism , Child , Child, Preschool , Cohort Studies , Craniopharyngioma/genetics , Craniopharyngioma/pathology , Cyst Fluid/immunology , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/physiology , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Male , Microarray Analysis , Pituitary Neoplasms/genetics , Pituitary Neoplasms/pathology , RNA, Messenger/metabolism , Receptors, Interleukin-6/genetics , Receptors, Interleukin-6/metabolism
17.
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
18.
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
19.
Pediatr Blood Cancer ; 64(11)2017 Nov.
Article in English | MEDLINE | ID: mdl-28383760

ABSTRACT

BACKGROUND: Nephrogenic rests (NRs) are abnormally persistent foci of embryonal cells, thought to be the precursor lesion of Wilms tumors (WTs). To date, their presence has not been systematically examined in WTs treated with preoperative chemotherapy. METHODS: A systematic analysis of the data on NRs in WTs treated with preoperative chemotherapy obtained from the UK cohort of the International Society of Pediatric Oncology (SIOP) WT 2001 Trial. The study was based on central pathology review of full sets of slides from pathological specimens, with a median of 28 slides reviewed per case. RESULTS: NRs were identified in 40% of unilateral WTs, including 25% perilobar nephrogenic rest (PLNR), 9% intralobar nephrogenic rest (ILNR), 5% both PLNR and ILNR, and 1% nephroblastomatosis, and in 93% of cases with bilateral lesions. ILNRs were associated with stromal histology and a younger age at diagnosis and found frequently in patients with congenital anomalies associated with WT1 mutation. PLNRs were found frequently in patients with overgrowth syndromes. CONCLUSIONS: The prevalence of NRs in WTs after preoperative chemotherapy observed in SIOP UK WT 2001 Trial is similar to the previously published data on NRs not treated with preoperative chemotherapy. Their epidemiology supports at least two pathways to Wilms tumorigenesis.


Subject(s)
Antineoplastic Agents/therapeutic use , Kidney Neoplasms/pathology , Wilms Tumor/pathology , Follow-Up Studies , Humans , International Agencies , Kidney Neoplasms/drug therapy , Kidney Neoplasms/epidemiology , Neoplasm Staging , Preoperative Care , Prevalence , Prognosis , United Kingdom/epidemiology , Wilms Tumor/drug therapy , Wilms Tumor/epidemiology
20.
Brain Pathol ; 27(3): 364-369, 2017 May.
Article in English | MEDLINE | ID: mdl-28414891

ABSTRACT

Adamantinomatous craniopharyngioma (ACP) is the commonest tumor of the sellar region in childhood. Two genetically engineered mouse models have been developed and are giving valuable insights into ACP biology. These models have identified novel pathways activated in tumors, revealed an important function of paracrine signalling and extended conventional theories about the role of organ-specific stem cells in tumorigenesis. In this review, we summarize these mouse models, what has been learnt, their limitations and open questions for future research. We then discussed how these mouse models may be used to test novel therapeutics against potentially targetable pathways recently identified in human ACP.


Subject(s)
Craniopharyngioma , Disease Models, Animal , Pituitary Neoplasms , Animals , Animals, Genetically Modified , Craniopharyngioma/drug therapy , Craniopharyngioma/genetics , Craniopharyngioma/physiopathology , Humans , Mice , Pituitary Neoplasms/genetics , Pituitary Neoplasms/physiopathology , Pituitary Neoplasms/therapy
SELECTION OF CITATIONS
SEARCH DETAIL
...