Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Neuropathol Appl Neurobiol ; 47(6): 781-795, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-33797808

RESUMEN

AIMS: We understand little of the pathogenesis of developmental cortical lesions, because we understand little of the diversity of the cell types that contribute to the diseases or how those cells interact. We tested the hypothesis that cellular diversity and cell-cell interactions play an important role in these disorders by investigating the signalling molecules in the commonest cortical malformations that lead to childhood epilepsy, focal cortical dysplasia (FCD) and tuberous sclerosis (TS). METHODS: Transcriptional profiling clustered cases into molecularly distinct groups. Using gene expression data, we identified the secretory signalling molecules in FCD/TS and characterised the cell types expressing these molecules. We developed a functional model using organotypic cultures. RESULTS: We identified 113 up-regulated secretory molecules in FCDIIB/TS. The top 12 differentially expressed genes (DEGs) were validated by immunohistochemistry. This highlighted two molecules, Chitinase 3-like protein 1 (CHI3L1) and C-C motif chemokine ligand 2 (CCL2) (MCP1) that were expressed in a unique population of small cells in close proximity to balloon cells (BC). We then characterised these cells and developed a functional model in organotypic slice cultures. We found that the number of CHI3L1 and CCL2 expressing cells decreased following inhibition of mTOR, the main aberrant signalling pathway in TS and FCD. CONCLUSIONS: Our findings highlight previously uncharacterised small cell populations in FCD and TS which express specific signalling molecules. These findings indicate a new level of diversity and cellular interactions in cortical malformations and provide a generalisable approach to understanding cell-cell interactions and cellular heterogeneity in developmental neuropathology.


Asunto(s)
Encéfalo/metabolismo , Discapacidades del Desarrollo/metabolismo , Malformaciones del Desarrollo Cortical/patología , Transducción de Señal/fisiología , Esclerosis Tuberosa/metabolismo , Encéfalo/patología , Discapacidades del Desarrollo/patología , Humanos , Inmunohistoquímica , Malformaciones del Desarrollo Cortical/metabolismo , Malformaciones del Desarrollo Cortical de Grupo I/metabolismo , Esclerosis Tuberosa/genética , Esclerosis Tuberosa/patología
2.
Cancer Discov ; 10(7): 942-963, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32238360

RESUMEN

Infant high-grade gliomas appear clinically distinct from their counterparts in older children, indicating that histopathologic grading may not accurately reflect the biology of these tumors. We have collected 241 cases under 4 years of age, and carried out histologic review, methylation profiling, and custom panel, genome, or exome sequencing. After excluding tumors representing other established entities or subgroups, we identified 130 cases to be part of an "intrinsic" spectrum of disease specific to the infant population. These included those with targetable MAPK alterations, and a large proportion of remaining cases harboring gene fusions targeting ALK (n = 31), NTRK1/2/3 (n = 21), ROS1 (n = 9), and MET (n = 4) as their driving alterations, with evidence of efficacy of targeted agents in the clinic. These data strongly support the concept that infant gliomas require a change in diagnostic practice and management. SIGNIFICANCE: Infant high-grade gliomas in the cerebral hemispheres comprise novel subgroups, with a prevalence of ALK, NTRK1/2/3, ROS1, or MET gene fusions. Kinase fusion-positive tumors have better outcome and respond to targeted therapy clinically. Other subgroups have poor outcome, with fusion-negative cases possibly representing an epigenetically driven pluripotent stem cell phenotype.See related commentary by Szulzewsky and Cimino, p. 904.This article is highlighted in the In This Issue feature, p. 890.


Asunto(s)
Fusión Génica/genética , Glioma/genética , Humanos , Lactante , Clasificación del Tumor , Pronóstico , Resultado del Tratamiento
3.
J Clin Invest ; 129(8): 3153-3170, 2019 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-31264977

RESUMEN

Hedgehog (Hh) proteins regulate development and tissue homeostasis, but their role in atopic dermatitis (AD) remains unknown. We found that on induction of mouse AD, Sonic Hedgehog (Shh) expression in skin, and Hh pathway action in skin T cells were increased. Shh signaling reduced AD pathology and the levels of Shh expression determined disease severity. Hh-mediated transcription in skin T cells in AD-induced mice increased Treg populations and their suppressive function through increased active transforming growth factor-ß (TGF-ß) in Tregs signaling to skin T effector populations to reduce disease progression and pathology. RNA sequencing of skin CD4+ T cells from AD-induced mice demonstrated that Hh signaling increased expression of immunoregulatory genes and reduced expression of inflammatory and chemokine genes. Addition of recombinant Shh to cultures of naive human CD4+ T cells in iTreg culture conditions increased FOXP3 expression. Our findings establish an important role for Shh upregulation in preventing AD, by increased Gli-driven Treg cell-mediated immune suppression, paving the way for a potential new therapeutic strategy.


Asunto(s)
Dermatitis Atópica/inmunología , Proteínas Hedgehog/inmunología , Transducción de Señal/inmunología , Piel/inmunología , Linfocitos T Reguladores/inmunología , Proteína Gli2 con Dedos de Zinc/inmunología , Animales , Dermatitis Atópica/genética , Dermatitis Atópica/patología , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/inmunología , Regulación de la Expresión Génica/inmunología , Proteínas Hedgehog/genética , Ratones , Ratones Noqueados , Transducción de Señal/genética , Piel/patología , Linfocitos T Reguladores/patología , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/inmunología , Proteína Gli2 con Dedos de Zinc/genética
4.
Hum Gene Ther ; 30(9): 1067-1078, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31288584

RESUMEN

Netherton syndrome (NS) is a rare autosomal recessive skin disorder caused by mutations in SPINK5. It is a debilitating condition with notable mortality in the early years of life. There is no curative treatment. We undertook a nonrandomized, open-label, feasibility, and safety study using autologous keratinocytes transduced with a lentiviral vector encoding SPINK5 under the control of the human involucrin promoter. Six NS subjects were recruited, and gene-modified epithelial sheets were successfully generated in three of five subjects. The sheets exhibited expression of correctly sized lympho-epithelial Kazal-type-related inhibitor (LEKTI) protein after modification. One subject was grafted with a 20 cm2 gene-modified graft on the left anterior thigh without any adverse complications and was monitored by serial sampling for 12 months. Recovery within the graft area was compared against an area outside by morphology, proviral copy number and expression of the SPINK5 encoded protein, LEKTI, and its downstream target kallikrein 5, which exhibited transient functional correction. The study confirmed the feasibility of generating lentiviral gene-modified epidermal sheets for inherited skin diseases such as NS, but sustained LEKTI expression is likely to require the identification, targeting, and engraftment of long-lived keratinocyte stem cell populations for durable therapeutic effects. Important learning points for the application of gene-modified epidermal sheets are discussed.


Asunto(s)
Células Epidérmicas/metabolismo , Epidermis/metabolismo , Epidermis/trasplante , Síndrome de Netherton/genética , Síndrome de Netherton/terapia , Transducción Genética , Transgenes , Adolescente , Adulto , Autoinjertos , Biomarcadores , Técnicas de Cultivo de Célula , Femenino , Técnica del Anticuerpo Fluorescente , Expresión Génica , Ingeniería Genética , Terapia Genética , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética , Humanos , Inmunohistoquímica , Queratinocitos/metabolismo , Lentivirus/genética , Masculino , Mutación , Síndrome de Netherton/metabolismo , Síndrome de Netherton/patología , Inhibidor de Serinpeptidasas Tipo Kazal-5/genética , Inhibidor de Serinpeptidasas Tipo Kazal-5/metabolismo , Resultado del Tratamiento , Adulto Joven
5.
Nat Commun ; 10(1): 2387, 2019 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-31160572

RESUMEN

Senescent cells accumulate in human tissues during ageing and contribute to age-related pathologies. The mechanisms responsible for their accumulation are unclear. Here we show that senescent dermal fibroblasts express the non-classical MHC molecule HLA-E, which interacts with the inhibitory receptor NKG2A expressed by NK and highly differentiated CD8+ T cells to inhibit immune responses against senescent cells. HLA-E expression is induced by senescence-associated secretary phenotype-related pro-inflammatory cytokines, and is regulated by p38 MAP kinase signalling in vitro. Consistently, HLA-E expression is increased on senescent cells in human skin sections from old individuals, when compared with those from young, and in human melanocytic nevi relative to normal skin. Lastly, blocking the interaction between HLA-E and NKG2A boosts immune responses against senescent cells in vitro. We thus propose that increased HLA-E expression contributes to persistence of senescent cells in tissues, thereby suggesting a new strategy for eliminating senescent cells during ageing.


Asunto(s)
Envejecimiento/inmunología , Linfocitos T CD8-positivos/inmunología , Senescencia Celular/inmunología , Fibroblastos/inmunología , Antígenos de Histocompatibilidad Clase I/inmunología , Células Asesinas Naturales/inmunología , Subfamília C de Receptores Similares a Lectina de Células NK/inmunología , Adulto , Anciano , Envejecimiento/patología , Citocinas/inmunología , Dermis/citología , Fibroblastos/patología , Humanos , Técnicas In Vitro , Nevo Pigmentado/congénito , Nevo Pigmentado/inmunología , Nevo Pigmentado/patología , Fenotipo , ARN Interferente Pequeño , Transducción de Señal , Piel/inmunología , Piel/patología , Adulto Joven , Proteínas Quinasas p38 Activadas por Mitógenos/inmunología , Antígenos HLA-E
6.
Hum Pathol ; 86: 182-192, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30594749

RESUMEN

The tumor suppressor TP53 promotes nerve growth factor receptor (NTRK1) -Y674/Y675 phosphorylation (NTRK1-pY674/pY675) via repression of the NTRK1 phosphatase PTPN6 in a ligand-independent manner, resulting in suppression of breast cancer cell proliferation. Moreover, NTRK1-pY674/pY675 together with low levels of PTPN6 and TP53 expression is associated with favorable disease-free survival of breast cancer patients. We determined whether in neuroblastoma this protein expression pattern impacts relapse-free survival (RFS). NTRK1-pY674/pY675, PTPN6, and TP53 expression was assessed in 98 neuroblastoma samples by immunohistochemistry. Association between expression levels and RFS was investigated by multivariate and Kaplan-Meier analysis. Mutant or wild-type TP53 was identified by sequencing tumor DNA. Tumors expressing NTRK1-pY674/pY675 and low or undetectable levels of PTPN6 and TP53 were significantly associated with 5-year RFS (P = .014) when the dataset was stratified by MYCN amplification, segmental chromosomal abnormalities and histology. Similar results were observed with tumors expressing wild-type TP53, NTRK1-pY674/pY675 and low or undetectable levels of PTPN6. Kaplan-Meier analysis demonstrated a significant correlation (P = .004), with a 50% probability of RFS (median survival 4.73 years) when present compared with 19.51% (median survival 11.63 months) when absent. Similar results were seen with non-amplified MYCN or unfavorable/undifferentiating samples and tumors from patients aged 18 months or less. Importantly, NTRK1-pY674/pY675 is an independent predictor of improved RFS. These results strongly suggest that NTRK1-pY674/pY675 together with wild-type TP53 and undetectable or low levels of PTPN6 expression is a potential biomarker of improved RFS of neuroblastoma patients. The predictive value of NTRK1-pY674/pY675 together with wild-type TP53 and low PTPN6 expression could contribute to neuroblastoma patient prognosis.


Asunto(s)
Neuroblastoma/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 6/metabolismo , Receptor trkA/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Adolescente , Biomarcadores de Tumor , Niño , Preescolar , Supervivencia sin Enfermedad , Regulación Neoplásica de la Expresión Génica , Humanos , Inmunohistoquímica , Lactante , Recién Nacido , Neuroblastoma/mortalidad , Neuroblastoma/patología , Fosforilación , Pronóstico , Tasa de Supervivencia
7.
F1000Res ; 8: 1544, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-32047602

RESUMEN

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.


Asunto(s)
Craneofaringioma , Neoplasias Hipofisarias , Craneofaringioma/diagnóstico , Craneofaringioma/historia , Historia del Siglo XIX , Historia del Siglo XX , Historia del Siglo XXI , Humanos , Imagen por Resonancia Magnética , Neoplasias Hipofisarias/diagnóstico , Neoplasias Hipofisarias/historia
8.
Circ Genom Precis Med ; 11(12): e001974, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30562113

RESUMEN

BACKGROUND: Hypertrophic cardiomyopathy (HCM) is characterized by a complex phenotype that is only partly explained by the biological effects of individual genetic variants. The aim of this study was to use proteomic analysis of myocardial tissue to explore the postgenomic phenotype. METHODS: Label-free proteomic analysis was used initially to compare protein profiles in myocardial samples from 11 patients with HCM undergoing surgical myectomy with control samples from 6 healthy unused donor hearts. Differentially expressed proteins of interest were validated in myocardial samples from 65 unrelated individuals (HCM [n=51], controls [n=7], and aortic stenosis [n=7]) by the development and use of targeted multiple reaction monitoring-based triple quadrupole mass spectrometry. RESULTS: In this exploratory study, 1586 proteins were identified with 151 proteins differentially expressed in HCM samples compared with controls ( P<0.05). Protein expression profiling showed that many proteins identified in the initial discovery study were associated with metabolism, muscle contraction, calcium regulation, and oxidative stress. Proteins downregulated in HCM versus controls included creatine kinase M-type, fructose-bisphosphate aldolase A, and phosphoglycerate mutase ( P<0.001). Proteins upregulated in HCM included lumican, carbonic anhydrase 3, desmin, α-actin skeletal, and FHL1 (four and a half LIM domain protein 1; P<0.01). Myocardial lumican concentration correlated with the left atrial area (ρ=0.34, P=0.015), late gadolinium enhancement on cardiac magnetic resonance imaging ( P=0.03) and the presence of a pathogenic sarcomere mutation ( P=0.04). CONCLUSIONS: The myocardial proteome of HCM provides supporting evidence for dysregulation of metabolic and structural proteins. The finding that lumican is raised in HCM hearts provides insight into the myocardial fibrosis that characterizes this disease.


Asunto(s)
Cardiomiopatía Hipertrófica/genética , Miocardio/metabolismo , Proteínas/química , Adulto , Anciano , Cardiomiopatía Hipertrófica/diagnóstico por imagen , Cardiomiopatía Hipertrófica/metabolismo , Femenino , Humanos , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Mutación , Miocardio/química , Fenotipo , Proteínas/genética , Proteínas/metabolismo , Proteómica
10.
Nat Commun ; 9(1): 4286, 2018 10 16.
Artículo en Inglés | MEDLINE | ID: mdl-30327457

RESUMEN

A tissue engineered oesophagus could overcome limitations associated with oesophageal substitution. Combining decellularized scaffolds with patient-derived cells shows promise for regeneration of tissue defects. In this proof-of-principle study, a two-stage approach for generation of a bio-artificial oesophageal graft addresses some major challenges in organ engineering, namely: (i) development of multi-strata tubular structures, (ii) appropriate re-population/maturation of constructs before transplantation, (iii) cryopreservation of bio-engineered organs and (iv) in vivo pre-vascularization. The graft comprises decellularized rat oesophagus homogeneously re-populated with mesoangioblasts and fibroblasts for the muscle layer. The oesophageal muscle reaches organised maturation after dynamic culture in a bioreactor and functional integration with neural crest stem cells. Grafts are pre-vascularised in vivo in the omentum prior to mucosa reconstitution with expanded epithelial progenitors. Overall, our optimised two-stage approach produces a fully re-populated, structurally organized and pre-vascularized oesophageal substitute, which could become an alternative to current oesophageal substitutes.


Asunto(s)
Esófago/citología , Esófago/fisiología , Músculo Esquelético/citología , Ingeniería de Tejidos/métodos , Andamios del Tejido , Animales , Técnicas de Cultivo de Célula , Diferenciación Celular , Niño , Preescolar , Criopreservación/métodos , Células Epiteliales , Matriz Extracelular/fisiología , Humanos , Lactante , Recién Nacido , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Cresta Neural/trasplante , Ratas Sprague-Dawley
11.
Mater Sci Eng C Mater Biol Appl ; 92: 565-574, 2018 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-30184783

RESUMEN

Cancer, disease and trauma to the larynx and their treatment can lead to permanent loss of structures critical to voice, breathing and swallowing. Engineered partial or total laryngeal replacements would need to match the ambitious specifications of replicating functionality, outer biocompatibility, and permissiveness for an inner mucosal lining. Here we present porous polyhedral oligomeric silsesquioxane-poly(carbonate urea) urethane (POSS-PCUU) as a potential scaffold for engineering laryngeal tissue. Specifically, we employ a precipitation and porogen leaching technique for manufacturing the polymer. The polymer is chemically consistent across all sample types and produces a foam-like scaffold with two distinct topographies and an internal structure composed of nano- and micro-pores. While the highly porous internal structure of the scaffold contributes to the complex tensile behaviour of the polymer, the surface of the scaffold remains largely non-porous. The low number of pores minimise access for cells, although primary fibroblasts and epithelial cells do attach and proliferate on the polymer surface. Our data show that with a change in manufacturing protocol to produce porous polymer surfaces, POSS-PCUU may be a potential candidate for overcoming some of the limitations associated with laryngeal reconstruction and regeneration.


Asunto(s)
Células Epiteliales/metabolismo , Fibroblastos/metabolismo , Laringe , Compuestos de Organosilicio/química , Poliuretanos/química , Ingeniería de Tejidos , Andamios del Tejido/química , Animales , Células Cultivadas , Células Epiteliales/citología , Fibroblastos/citología , Porcinos
12.
Nat Commun ; 9(1): 3505, 2018 08 29.
Artículo en Inglés | MEDLINE | ID: mdl-30158522

RESUMEN

Argininosuccinate lyase (ASL) belongs to the hepatic urea cycle detoxifying ammonia, and the citrulline-nitric oxide (NO) cycle producing NO. ASL-deficient patients present argininosuccinic aciduria characterised by hyperammonaemia, multiorgan disease and neurocognitive impairment despite treatment aiming to normalise ammonaemia without considering NO imbalance. Here we show that cerebral disease in argininosuccinic aciduria involves neuronal oxidative/nitrosative stress independent of hyperammonaemia. Intravenous injection of AAV8 vector into adult or neonatal ASL-deficient mice demonstrates long-term correction of the hepatic urea cycle and the cerebral citrulline-NO cycle, respectively. Cerebral disease persists if ammonaemia only is normalised but is dramatically reduced after correction of both ammonaemia and neuronal ASL activity. This correlates with behavioural improvement and reduced cortical cell death. Thus, neuronal oxidative/nitrosative stress is a distinct pathophysiological mechanism from hyperammonaemia. Disease amelioration by simultaneous brain and liver gene transfer with one vector, to treat both metabolic pathways, provides new hope for hepatocerebral metabolic diseases.


Asunto(s)
Argininosuccinatoliasa/metabolismo , Aciduria Argininosuccínica/metabolismo , Aciduria Argininosuccínica/terapia , Animales , Argininosuccinatoliasa/genética , Aciduria Argininosuccínica/genética , Encefalopatías/genética , Encefalopatías/metabolismo , Encefalopatías/terapia , Citrulina/metabolismo , Terapia Genética , Hiperamonemia/genética , Hiperamonemia/metabolismo , Hiperamonemia/terapia , Hígado/citología , Ratones , Neuronas/metabolismo , Óxido Nítrico/metabolismo , Estrés Nitrosativo/genética , Estrés Nitrosativo/fisiología
13.
Acta Neuropathol ; 135(5): 757-777, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29541918

RESUMEN

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.


Asunto(s)
Craneofaringioma/metabolismo , Sistema de Señalización de MAP Quinasas , Neoplasias Hipofisarias/metabolismo , Transcriptoma , Microambiente Tumoral/fisiología , Animales , Biología Computacional , Craneofaringioma/patología , Craneofaringioma/terapia , Citocinas/metabolismo , Modelos Animales de Enfermedad , Humanos , Inflamación/metabolismo , Inflamación/terapia , Captura por Microdisección con Láser , Ratones , Neuroglía/metabolismo , Odontogénesis/fisiología , Hipófisis/embriología , Hipófisis/patología , Neoplasias Hipofisarias/patología , Neoplasias Hipofisarias/terapia , Análisis de Secuencia de ARN , Técnicas de Cultivo de Tejidos
14.
Kidney Int ; 93(4): 903-920, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29398135

RESUMEN

The Wilms' tumor suppressor gene, WT1, encodes a zinc finger protein that regulates podocyte development and is highly expressed in mature podocytes. Mutations in the WT1 gene are associated with the development of renal failure due to the formation of scar tissue within glomeruli, the mechanisms of which are poorly understood. Here, we used a tamoxifen-based CRE-LoxP system to induce deletion of Wt1 in adult mice to investigate the mechanisms underlying evolution of glomerulosclerosis. Podocyte apoptosis was evident as early as the fourth day post-induction and increased during disease progression, supporting a role for Wt1 in mature podocyte survival. Podocyte Notch activation was evident at disease onset with upregulation of Notch1 and its transcriptional targets, including Nrarp. There was repression of podocyte FoxC2 and upregulation of Hey2 supporting a role for a Wt1/FoxC2/Notch transcriptional network in mature podocyte injury. The expression of cleaved Notch1 and HES1 proteins in podocytes of mutant mice was confirmed in early disease. Furthermore, induction of podocyte HES1 expression was associated with upregulation of genes implicated in epithelial mesenchymal transition, thereby suggesting that HES1 mediates podocyte EMT. Lastly, early pharmacological inhibition of Notch signaling ameliorated glomerular scarring and albuminuria. Thus, loss of Wt1 in mature podocytes modulates podocyte Notch activation, which could mediate early events in WT1-related glomerulosclerosis.


Asunto(s)
Glomerulonefritis/metabolismo , Podocitos/metabolismo , Receptor Notch1/metabolismo , Proteínas Represoras/metabolismo , Albuminuria/genética , Albuminuria/metabolismo , Animales , Apoptosis , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Células Cultivadas , Modelos Animales de Enfermedad , Transición Epitelial-Mesenquimal , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Regulación de la Expresión Génica , Glomerulonefritis/genética , Glomerulonefritis/patología , Péptidos y Proteínas de Señalización Intracelular , Ratones Endogámicos C57BL , Ratones Noqueados , Podocitos/patología , Proteínas/genética , Proteínas/metabolismo , Receptor Notch1/genética , Proteínas Represoras/deficiencia , Proteínas Represoras/genética , Transducción de Señal , Transcripción Genética , Proteínas WT1
15.
J Clin Invest ; 128(4): 1496-1508, 2018 04 02.
Artículo en Inglés | MEDLINE | ID: mdl-29461977

RESUMEN

BACKGROUND: Sporadic vascular malformations (VMs) are complex congenital anomalies of blood vessels that lead to stroke, life-threatening bleeds, disfigurement, overgrowth, and/or pain. Therapeutic options are severely limited, and multidisciplinary management remains challenging, particularly for high-flow arteriovenous malformations (AVM). METHODS: To investigate the pathogenesis of sporadic intracranial and extracranial VMs in 160 children in which known genetic causes had been excluded, we sequenced DNA from affected tissue and optimized analysis for detection of low mutant allele frequency. RESULTS: We discovered multiple mosaic-activating variants in 4 genes of the RAS/MAPK pathway, KRAS, NRAS, BRAF, and MAP2K1, a pathway commonly activated in cancer and responsible for the germline RAS-opathies. These variants were more frequent in high-flow than low-flow VMs. In vitro characterization and 2 transgenic zebrafish AVM models that recapitulated the human phenotype validated the pathogenesis of the mutant alleles. Importantly, treatment of AVM-BRAF mutant zebrafish with the BRAF inhibitor vemurafinib restored blood flow in AVM. CONCLUSION: Our findings uncover a major cause of sporadic VMs of different clinical types and thereby offer the potential of personalized medical treatment by repurposing existing licensed cancer therapies. FUNDING: This work was funded or supported by grants from the AVM Butterfly Charity, the Wellcome Trust (UK), the Medical Research Council (UK), the UK National Institute for Health Research, the L'Oreal-Melanoma Research Alliance, the European Research Council, and the National Human Genome Research Institute (US).


Asunto(s)
Alelos , MAP Quinasa Quinasa 1 , Sistema de Señalización de MAP Quinasas/genética , Mutación , Fenotipo , Malformaciones Vasculares , Proteínas ras , Adolescente , Adulto , Animales , Niño , Femenino , Células HEK293 , Células Endoteliales de la Vena Umbilical Humana , Humanos , Lactante , MAP Quinasa Quinasa 1/genética , MAP Quinasa Quinasa 1/metabolismo , Masculino , Malformaciones Vasculares/genética , Malformaciones Vasculares/metabolismo , Malformaciones Vasculares/patología , Pez Cebra , Proteínas ras/genética , Proteínas ras/metabolismo
16.
Brain Pathol ; 28(4): 475-483, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-28481062

RESUMEN

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.


Asunto(s)
Craneofaringioma/diagnóstico por imagen , Craneofaringioma/patología , Modelos Animales de Enfermedad , Animales , Craneofaringioma/genética , Xenoinjertos/diagnóstico por imagen , Xenoinjertos/patología , Proteínas de Homeodominio/genética , Humanos , Imagen por Resonancia Magnética , Masculino , Ratones Transgénicos , Persona de Mediana Edad , Proteínas Represoras/genética , Microtomografía por Rayos X , beta Catenina/genética
17.
Acta Neuropathol ; 135(1): 115-129, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29058119

RESUMEN

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.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Epilepsia/metabolismo , Ganglioglioma/metabolismo , Neoplasias Neuroepiteliales/metabolismo , Adolescente , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/cirugía , Niño , Preescolar , Estudios de Cohortes , Metilación de ADN , Epilepsia/genética , Epilepsia/patología , Epilepsia/cirugía , Femenino , Ganglioglioma/genética , Ganglioglioma/patología , Ganglioglioma/cirugía , Expresión Génica , Humanos , Lactante , Masculino , Mutación , Neoplasias Neuroepiteliales/genética , Neoplasias Neuroepiteliales/patología , Neoplasias Neuroepiteliales/cirugía , Fenotipo , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas B-raf/metabolismo , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo
18.
Placenta ; 60: 36-39, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29208237

RESUMEN

Micro-CT provides 3D volume imaging with spatial resolution at the micrometre scale. We investigated the optimal human placenta tissue preparation (contrast agent, perfusion pressure, perfusion location and perfusion vessel) and imaging (energy, target material, exposure time and frames) parameters. Microfil (Flow Tech, Carver, MA) produced better fill than Barium sulphate (84.1%(±11.5%)vs70.4%(±18.02%) p = 0.01). Perfusion via umbilical artery produced better fill than via chorionic vessels (83.8%(±17.7%)vs78.0%(±21.9%), p < 0.05), or via umbilical vein (83.8%(±16.4%)vs69.8%(±20.3%), p < 0.01). Imaging at 50 keV with a molybdenum target produced the best contrast to noise ratio. We propose this method to enable quantification and comparison of the human fetoplacental vascular tree.


Asunto(s)
Placenta/diagnóstico por imagen , Microtomografía por Rayos X/métodos , Sulfato de Bario , Femenino , Humanos , Microcirculación , Perfusión , Placenta/irrigación sanguínea , Embarazo , Elastómeros de Silicona
19.
Nat Commun ; 8(1): 1819, 2017 11 28.
Artículo en Inglés | MEDLINE | ID: mdl-29180744

RESUMEN

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.


Asunto(s)
Senescencia Celular/fisiología , Craneofaringioma/metabolismo , Células Madre Neoplásicas/metabolismo , Neoplasias Hipofisarias/metabolismo , Compuestos de Anilina/farmacología , Animales , Compuestos de Bifenilo/farmacología , Transformación Celular Neoplásica , Niño , Craneofaringioma/patología , Modelos Animales de Enfermedad , Proteínas de Homeodominio/metabolismo , Humanos , Ratones , Nitrofenoles/farmacología , Oncogenes/fisiología , Piperazinas/farmacología , Hipófisis/metabolismo , Hipófisis/patología , Neoplasias Hipofisarias/patología , Proteínas Represoras/metabolismo , Factores de Transcripción SOXB1/metabolismo , Sulfonamidas/farmacología , Secuenciación del Exoma , Adulto Joven , beta Catenina/metabolismo
20.
Placenta ; 59: 69-72, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-29108639

RESUMEN

Proteomic characterisation of the placenta has largely been focused on effect of disease, anatomical features or specific cell types. We describe an unbiased proteomic mapping analysis to investigate how the placental proteome changes throughout the organ. A transverse slice of a human placenta was sectioned into 1 × 1cm samples. Sections were analysed using label free proteomics. Analysis revealed two distinct sub-proteomes that did not have anatomical significance. One had a muscular proteome and the other had distinct immunomodulation functions. Chorionic plate enriched proteins highlighted the fetal tissues high energy requirements whilst mechanisms of the decidua observed included modulation of cortisone levels.


Asunto(s)
Placenta/metabolismo , Proteoma , Femenino , Humanos , Embarazo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...