Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
2.
Am J Respir Crit Care Med ; 205(3): 300-312, 2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-34860143

RESUMEN

Rationale: The alarmins IL-33 and HMGB1 (high mobility group box 1) contribute to type 2 inflammation and asthma pathogenesis. Objectives: To determine whether P2Y13-R (P2Y13 receptor), a purinergic GPCR (G protein-coupled receptor) and risk allele for asthma, regulates the release of IL-33 and HMGB1. Methods: Bronchial biopsy specimens were obtained from healthy subjects and subjects with asthma. Primary human airway epithelial cells (AECs), primary mouse AECs, or C57Bl/6 mice were inoculated with various aeroallergens or respiratory viruses, and the nuclear-to-cytoplasmic translocation and release of alarmins was measured by using immunohistochemistry and an ELISA. The role of P2Y13-R in AEC function and in the onset, progression, and exacerbation of experimental asthma was assessed by using pharmacological antagonists and mice with P2Y13-R gene deletion. Measurements and Main Results: Aeroallergen exposure induced the extracellular release of ADP and ATP, nucleotides that activate P2Y13-R. ATP, ADP, and aeroallergen (house dust mite, cockroach, or Alternaria antigen) or virus exposure induced the nuclear-to-cytoplasmic translocation and subsequent release of IL-33 and HMGB1, and this response was ablated by genetic deletion or pharmacological antagonism of P2Y13. In mice, prophylactic or therapeutic P2Y13-R blockade attenuated asthma onset and, critically, ablated the severity of a rhinovirus-associated exacerbation in a high-fidelity experimental model of chronic asthma. Moreover, P2Y13-R antagonism derepressed antiviral immunity, increasing IFN-λ production and decreasing viral copies in the lung. Conclusions: We identify P2Y13-R as a novel gatekeeper of the nuclear alarmins IL-33 and HMGB1 and demonstrate that the targeting of this GPCR via genetic deletion or treatment with a small-molecule antagonist protects against the onset and exacerbations of experimental asthma.


Asunto(s)
Asma/inmunología , Proteína HMGB1/metabolismo , Interleucina-33/metabolismo , Receptores Purinérgicos P2/metabolismo , Animales , Asma/metabolismo , Asma/fisiopatología , Biomarcadores/metabolismo , Estudios de Casos y Controles , Progresión de la Enfermedad , Ensayo de Inmunoadsorción Enzimática , Células Epiteliales/metabolismo , Humanos , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL
3.
Exp Dermatol ; 29(5): 450-461, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32145039

RESUMEN

Mouse dorsal coat hair types, guard, awl, auchene and zigzag, develop in three consecutive waves. To date, it is unclear if these hair types are determined genetically through expression of specific factors or can change based on their mesenchymal environment. We undertook a novel approach to this question by studying individual hair type in 67 Collaborative Cross (CC) mouse lines and found significant variation in the proportion of each type between strains. Variation in the proportion of zigzag, awl and auchene, but not guard hair, was largely due to germline genetic variation. We utilised this variation to map a quantitative trait locus (QTL) on chromosome 12 that appears to influence a decision point switch controlling the propensity for either second (awl and auchene) or third wave (zigzag) hairs to develop. This locus contains two strong candidates, Sostdc1 and Twist1, each of which carry several ENCODE regulatory variants, specific to the causal allele, that can influence gene expression, are expressed in the developing hair follicle, and have been previously reported to be involved in regulating human and murine hair behaviour, but not hair subtype determination. Both of these genes are likely to play a part in hair type determination via regulation of BMP and/or WNT signalling.


Asunto(s)
Proteínas Morfogenéticas Óseas/metabolismo , Mapeo Cromosómico , Estudio de Asociación del Genoma Completo , Cabello/fisiología , Polimorfismo Genético , Transducción de Señal , Proteínas Wnt/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Cruzamientos Genéticos , Dermis/metabolismo , Ligamiento Genético , Ratones , Fenotipo , Sitios de Carácter Cuantitativo , Especificidad de la Especie , Proteína 1 Relacionada con Twist/genética
5.
Elife ; 82019 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-30681412

RESUMEN

Genetic variation conferring resistance and susceptibility to carcinogen-induced tumorigenesis is frequently studied in mice. We have now turned this idea to melanoma using the collaborative cross (CC), a resource of mouse strains designed to discover genes for complex diseases. We studied melanoma-prone transgenic progeny across seventy CC genetic backgrounds. We mapped a strong quantitative trait locus for rapid onset spontaneous melanoma onset to Prkdc, a gene involved in detection and repair of DNA damage. In contrast, rapid onset UVR-induced melanoma was linked to the ribosomal subunit gene Rrp15. Ribosome biogenesis was upregulated in skin shortly after UVR exposure. Mechanistically, variation in the 'usual suspects' by which UVR may exacerbate melanoma, defective DNA repair, melanocyte proliferation, or inflammatory cell infiltration, did not explain melanoma susceptibility or resistance across the CC. Instead, events occurring soon after exposure, such as dysregulation of ribosome function, which alters many aspects of cellular metabolism, may be important.


Asunto(s)
Melanoma/genética , Neoplasias Cutáneas/genética , Rayos Ultravioleta , Animales , Animales Recién Nacidos , Animales no Consanguíneos , Proliferación Celular , Mapeo Cromosómico , Cromosomas de los Mamíferos/genética , Modelos Animales de Enfermedad , Regulación Neoplásica de la Expresión Génica , Redes Reguladoras de Genes , Sitios Genéticos , Humanos , Melanocitos/metabolismo , Melanocitos/patología , Melanoma/patología , Ratones Transgénicos , Proteínas de Unión al GTP Monoméricas/genética , Proteínas Proto-Oncogénicas B-raf/genética , Sitios de Carácter Cuantitativo/genética , Reproducibilidad de los Resultados , Piel/metabolismo , Piel/patología , Neoplasias Cutáneas/patología , Melanoma Cutáneo Maligno
6.
J Invest Dermatol ; 139(1): 177-185, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30009829

RESUMEN

Melanocytes can group together in nevi, commonly thought to form because of intrinsic somatic mutations involving MAPK pathway activation. However, the role of the microenvironment, in particular keratinocytes, in nevogenesis is rarely studied. Melanocytes proliferate during the hair follicle growth phase and in some basal cell carcinomas, allowing us to construct keratinocyte gene expression clusters correlated with melanocyte activation. We asked whether such correlations are evident in the more subtle context of regulation of melanocyte behavior in normal skin. We considered genes which, when mutated in keratinocytes in mice, lead to nevogenesis. Across the human GTEx normal skin database, their expression was correlated with that of keratinocyte cytokines KITLG, HGF, FGF2, EDN1, and melanocyte markers. These cytokines have pleiotropic effects on melanocyte-specific and pigmentation genes and also influence mast cell gene expression. We show five classes of keratinocyte genes that, via germline genetic variation, influence melanocyte activity. These include genes involved in SHH signaling, structural keratins, ribosomal biogenesis, and stem cell governance. In agreement with the finding of KITLG linked to nevogenesis in human genome-wide association studies, we provide evidence that specific keratinocyte cytokines are components of networks that may drive or exacerbate nevus development.


Asunto(s)
Citocinas/genética , Regulación Neoplásica de la Expresión Génica , Queratinocitos/metabolismo , Nevo Pigmentado/genética , ARN Neoplásico/genética , Neoplasias Cutáneas/genética , Animales , Citocinas/biosíntesis , Estudio de Asociación del Genoma Completo , Humanos , Queratinocitos/patología , Ratones , Ratones Noqueados , Nevo Pigmentado/metabolismo , Nevo Pigmentado/patología , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/patología
7.
J Invest Dermatol ; 138(4): 893-902, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29138054

RESUMEN

Giant congenital nevi are associated with clinical complications such as neurocutaneous melanosis and melanoma. Virtually nothing is known about why some individuals develop these lesions. We previously identified the sonic hedgehog (Shh) pathway regulator Cdon as a candidate nevus modifier gene. Here we validate this by studying Cdon knockout mice, and go on to establishing the mechanism by which Shh exacerbates nevogenesis. Cdon knockout mice develop blue nevi without the need for somatic melanocyte oncogenic mutation. In a mouse model carrying melanocyte NRASQ61K, we found that strain backgrounds that carry genetic variants that cause increased keratinocyte Shh pathway activity, as measured by Gli1 and Gli2 expression, develop giant congenital nevi. Shh components are also active adjacent to human congenital nevi. Mechanistically, this exacerbation of nevogenesis is driven via the release of the melanocyte mitogen endothelin-1 from keratinocytes. We then suppressed nevus development in mice using Shh and endothelin antagonists. Our work suggests an aspect of nevus development whereby keratinocyte cytokines such as endothelin-1 can exacerbate nevogenesis, and provides potential therapeutic approaches for giant congenital nevi. Furthermore, it highlights the notion that germline genetic variation, in addition to somatic melanocyte mutation, can strongly influence the histopathological features of melanocytic nevi.


Asunto(s)
Endotelina-1/metabolismo , Regulación Neoplásica de la Expresión Génica , Proteínas Hedgehog/genética , Queratinocitos/metabolismo , Neoplasias Experimentales , Nevo Pigmentado/genética , Neoplasias Cutáneas/genética , Regulación hacia Arriba , Animales , Femenino , Proteínas Hedgehog/biosíntesis , Humanos , Queratinocitos/patología , Masculino , Melanocitos/metabolismo , Melanocitos/patología , Ratones , Ratones Noqueados , Nevo Pigmentado/metabolismo , Nevo Pigmentado/patología , Transducción de Señal , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/patología , Activación Transcripcional , Células Tumorales Cultivadas
8.
Pigment Cell Melanoma Res ; 29(4): 459-64, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27155367

RESUMEN

Congenital nevi develop before birth and sometimes cover large areas of the body. They are presumed to arise from the acquisition of a gene mutation in an embryonic melanocyte that becomes trapped in the dermis during development. Mice bearing the Cdk4(R24C) ::Tyr-NRAS(Q) (61K) transgenes develop congenital nevus-like lesions by post-natal day 10, from melanocytes escaping the confines of hair follicles. We interbred these mice with the collaborative cross (CC), a resource that enables identification of modifier genes for complex diseases (those where multiple genes are involved). We examined variation in nevus cell density in 66 CC strains and mapped a large-effect quantitative trait locus (QTL) controlling nevus cell density to murine chromosome 9. The best candidate for a gene that exacerbates congenital nevus development in the context of an NRAS mutation is Cdon, a positive regulator of sonic hedgehog (Shh) that is expressed mainly in keratinocytes.


Asunto(s)
Moléculas de Adhesión Celular/genética , GTP Fosfohidrolasas/genética , Melanocitos/patología , Proteínas de la Membrana/genética , Mutación , Nevo/congénito , Neoplasias Cutáneas/congénito , Animales , Células Cultivadas , Dermis/metabolismo , Dermis/patología , Femenino , Folículo Piloso/metabolismo , Folículo Piloso/patología , Humanos , Queratinocitos/metabolismo , Queratinocitos/patología , Masculino , Melanocitos/metabolismo , Ratones , Ratones Noqueados , Nevo/patología , Neoplasias Cutáneas/patología
10.
Photochem Photobiol Sci ; 14(6): 1179-89, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25966309

RESUMEN

Melanocyte stem cells (MCSCs) in the upper portion of the hair follicle periodically supply melanocytes (MCs) that migrate downward into the hair bulb during anagen, the growth phase of the hair cycle. However MCs can also migrate upwards. We previously observed an increase in epidermal MC density in the mouse epidermis after a single ultraviolet radiation (UVR) exposure in neonatal, but not adult mice. To better understand MCSC activation by UVR we methodically studied the response of MCs to narrow band UVB (since UVA does not invoke this response) exposure in neonatal mice, and in adults at different stages of the hair cycle. We found that a single exposure of adult mice did not induce activation of MCSCs, in any stage of the hair cycle. When adult mice MCSCs were isolated in telogen, multiple UVB exposures resulted in their activation and production of daughter cells, which migrated upwards to the epidermis. Importantly, the MCSCs produced new progeny without themselves having incurred DNA damage after UVB exposure. This, together with examination of MC localisation in the skin of mice overexpressing stem cell factor in their keratinocytes, leads us to conclude that MCSC activation by UVB is driven via paracrine production of either SCF and/or other keratinocyte cytokines. We re-examined the increase in epidermal MC density in neonatal mouse skin. This effect was much more profound after only a single exposure than that of even multiple exposures to adult skin, and we show that in this setting also, the epidermal MCs mostly derive from activation of MC precursors in the upper hair follicle, and most likely via a cell extrinsic mechanism. Hence, although adaptive changes in the skin induced by repetitive UVB exposures are necessary in adult mice, in both the adult and neonatal context the division and migration upwards of follicular MCSCs is the major mode by which epidermal MC numbers increase after UVR exposure.


Asunto(s)
Folículo Piloso/citología , Folículo Piloso/efectos de la radiación , Melanocitos/efectos de la radiación , Rayos Ultravioleta , Animales , Proliferación Celular/efectos de la radiación , Daño del ADN , Inmunohistoquímica , Melanocitos/citología , Ratones , Piel/citología , Piel/efectos de la radiación , Células Madre/citología , Células Madre/efectos de la radiación
11.
Curr Probl Dermatol ; 46: 42-8, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25561205

RESUMEN

This manuscript focuses on the use of mice to study the genetics and biology of cutaneous squamous cell carcinoma (SCC). Mice develop actinic keratosis-like lesions and SCC resembling those seen in humans. As an animal model, the mouse provides great experimental flexibility and has been useful in investigating aspects of the genetics and biology of SCC that are difficult to study in humans. We discuss the pros and cons of the various murine models available. How well mouse pathology in general mimics human disease remains an open question due to the vast differences in animal strain backgrounds and the fact that only one strain is typically tested in any particular experiment. Nonetheless, the murine epidermis is thinner than the human epidermis, and this must be kept in mind when making inferences from mechanistic data obtained with mice. We outline new strategies for non-biased screens to discover genes driving SCC progression. Such work has revealed a very complex interactive molecular network, and as with other complex diseases, the picture is being pieced together using systems biology strategies to which mouse tumour models are amenable. Such approaches do not focus on single genes or proteins but try to integrate the complex interactions of many types of genetic and biological information.


Asunto(s)
Carcinoma de Células Escamosas , Modelos Animales de Enfermedad , Queratosis Actínica , Neoplasias Experimentales , Neoplasias Cutáneas , Animales , Carcinoma de Células Escamosas/inducido químicamente , Carcinoma de Células Escamosas/genética , Queratosis Actínica/inducido químicamente , Queratosis Actínica/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neoplasias Experimentales/inducido químicamente , Neoplasias Experimentales/genética , Neoplasias Cutáneas/inducido químicamente , Neoplasias Cutáneas/genética
12.
J Invest Dermatol ; 134(7): 1991-1997, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24509534

RESUMEN

Epidemiological studies suggest that ultraviolet B exposure (UVR) during childhood is the most important environmental risk factor for melanoma. In accordance, neonatal, but not adult, UVR exacerbates melanoma incidence in mouse models. The inability of neonates, as opposed to adults, to mount a proper neutrophil inflammatory response in the skin upon UVR exposure has been one of the driving hypotheses explaining this observation for the past decade. However, this aspect remains controversial. Here, we evaluated the UVR-induced inflammatory response in neonatal versus adult mice. In neonates, a significant neutrophil infiltration could be identified and quantified using three different antibodies by flow cytometry or immunohistochemistry. On day 1 after UVR, neutrophils were increased by 84-fold and on day 4 macrophages increased by 37-fold compared with nonexposed age-matched skin. When compared with adults, neonatal skin harbored a higher proportion of neutrophils in the myeloid compartment without significant differences in absolute counts. This response was reproduced with different kinetics in C57Bl/6 and FVB mice with a more rapid attenuation of neutrophil counts in the latter. Overall, our results suggest that the greatly increased sensitivity to melanomagenesis in neonates does not result from their incompetence in terms of myeloid inflammatory response to UVR.


Asunto(s)
Macrófagos/patología , Melanoma/patología , Neoplasias Inducidas por Radiación/patología , Neutrófilos/patología , Neoplasias Cutáneas/patología , Rayos Ultravioleta/efectos adversos , Factores de Edad , Animales , Animales Recién Nacidos , Proliferación Celular/efectos de la radiación , Citometría de Flujo , Macrófagos/efectos de la radiación , Melanoma/epidemiología , Melanoma/inmunología , Ratones , Ratones Endogámicos C57BL , Células Mieloides/patología , Células Mieloides/efectos de la radiación , Neoplasias Inducidas por Radiación/epidemiología , Neoplasias Inducidas por Radiación/inmunología , Neutrófilos/efectos de la radiación , Factores de Riesgo , Neoplasias Cutáneas/epidemiología , Neoplasias Cutáneas/inmunología
13.
Pigment Cell Melanoma Res ; 26(5): 731-4, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23724991

RESUMEN

We previously noted that melanomas developing in Cdk4(R24C/R24C) ::Tyr-NRAS, Arf(-/-) ::Tyr-NRAS and Trp53(F/F) ::Tyr-Cre(ER)::Tyr-NRAS mice exhibited differences in behaviour in vivo. We investigated this phenomenon using global gene expression profiling of lesions from the respective genotypes. While those from the Cdk4- and Arf-mutant mice exhibited similar profiles, the Trp53(F/F) ::Tyr-Cre(ER)::Tyr-NRAS melanomas were strikingly different, showing relative down-regulation of melanocyte-related genes, and up-regulation of genes related to neural differentiation. Specifically, they highly expressed genes representative of the myelin-producing peripheral oligodendrite (Schwann cell) lineage, although histopathologically the lesions did not exhibit the classical features of schwannoma. As Schwann cell precursors can be a cellular origin of melanocytes, it is unsurprising that plasticity with respect to melanocyte-neural differentiation can occur in melanoma. What is surprising is the genotype proclivity. Comparison of gene expression signatures revealed that melanomas from the Trp53-mutant mice show significant similarities with a subset of aggressive human melanomas with relatively low levels of MITF.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Transdiferenciación Celular , Quinasa 4 Dependiente de la Ciclina/metabolismo , Melanoma/metabolismo , Melanoma/patología , Neuronas/patología , Proteolisis , Proteína p53 Supresora de Tumor/metabolismo , Animales , Transdiferenciación Celular/genética , Quinasa 4 Dependiente de la Ciclina/genética , Regulación hacia Abajo , Regulación Neoplásica de la Expresión Génica , Humanos , Melanoma/genética , Ratones , Neuronas/metabolismo , Regulación hacia Arriba/genética
14.
J Invest Dermatol ; 133(7): 1803-12, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23321920

RESUMEN

Intermittent sunburns, particularly in childhood, are the strongest environmental risk factor for malignant melanoma (MM). In mice, a single neonatal UVR exposure induces MM, whereas chronic doses to adult mice do not. Neonatal UVR alters melanocyte migration dynamics by inducing their movement upward out of hair follicles into the epidermis. UVR is known to induce inflammation and recruitment of macrophages into the skin. In this study, we have used a liposomal clodronate strategy to deplete macrophages at the time of neonatal UVR, and have shown functionally that this reduces the melanocyte proliferative response. This effect was not reproduced by depletion of CD11c-expressing populations of dendritic cells. On the basis of epidermal expression array data at various time points after UVR, we selected mouse strains defective in various aspects of macrophage recruitment, activation, and effector functions, and measured their melanocyte UVR response. We identified Ly6c(low)MHCII(hi) macrophages as the major population promoting the melanocyte response across multiple strains. The activity of this subpopulation was CCR2 (C-C chemokine receptor type 2) independent and partly IL-17 dependent. By helping induce this effect, the infiltration of specific macrophage subpopulations after sunburn may be a factor in increasing the risk of subsequent neoplastic transformation of melanocytes.


Asunto(s)
Antígenos Ly/metabolismo , Proliferación Celular/efectos de la radiación , Antígenos de Histocompatibilidad Clase II/metabolismo , Macrófagos/inmunología , Melanocitos/metabolismo , Receptores CCR2/metabolismo , Rayos Ultravioleta , Animales , Animales Recién Nacidos , Transformación Celular Neoplásica/patología , Interleucina-17/metabolismo , Macrófagos/patología , Melanocitos/patología , Melanocitos/efectos de la radiación , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Receptores CCR2/deficiencia , Receptores CCR2/genética , Factores de Riesgo , Piel/metabolismo , Piel/patología , Quemadura Solar/complicaciones , Factores de Tiempo
17.
Pigment Cell Melanoma Res ; 23(6): 771-80, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20718941

RESUMEN

We report on a systematic analysis of genotype-specific melanocyte (MC) UVR responses in transgenic mouse melanoma models along with tumour penetrance and comparative histopathology. pRb or p53 pathway mutations cooperated with Nras(Q61K) to transform MCs. We previously reported that MCs migrate from the follicular outer root sheath into the epidermis after neonatal UVR. Here, we found that Arf or p53 loss markedly diminished this response. Despite this, mice carrying these mutations developed melanoma with very early age of onset after neonatal UVR. Cdk4(R24C) did not affect the MC migration. Instead, independent of UVR exposure, interfollicular dermal MCs were more prevalent in Cdk4(R24C) mice. Subsequently, in adulthood, these mutants developed dermal MC proliferations reminiscent of superficial congenital naevi. Two types of melanoma were observed in this model. The location and growth pattern of the first was consistent with derivation from the naevi, while the second appeared to be of deep dermal origin. In animals carrying the Arf or p53 defects, no naevi were detected, with all tumours ostensibly skipping the benign precursor stage in progression.


Asunto(s)
Melanoma/metabolismo , Nevo/metabolismo , Lesiones Precancerosas/patología , Proteína de Retinoblastoma/metabolismo , Transducción de Señal , Proteína p14ARF Supresora de Tumor/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Animales , Animales Recién Nacidos , Recuento de Células , Movimiento Celular/efectos de la radiación , Proliferación Celular/efectos de la radiación , Quinasa 4 Dependiente de la Ciclina/metabolismo , Melanocitos/patología , Melanocitos/efectos de la radiación , Melanoma/genética , Melanoma/patología , Ratones , Modelos Biológicos , Nevo/genética , Nevo/patología , Penetrancia , Lesiones Precancerosas/metabolismo , Transducción de Señal/efectos de la radiación , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/patología , Rayos Ultravioleta
18.
Psychiatr Genet ; 18(5): 219-25, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18797396

RESUMEN

OBJECTIVES: Association of the valine/methionine variant at codon 66 (Val66Met) of brain derived neurotrophic factor (BDNF) has been reported inconsistently across a spectrum of psychiatric disorders. Haplotypes of six tagging single nucleotide polymorphisms (SNPs) of a 37-kb region of dystrobrevin-binding protein 1 (DTNBP1) were found to be associated with schizophrenia. These haplotypes have not been studied extensively for other psychiatric disorders but are plausible candidates for anxiety and depression disorders. Here, association between variants of BDNF and DTNBP1, and multiple anxiety and depression phenotypes is explored. METHODS: Study participants were selected as sibling pairs that were either concordant or discordant for extreme neuroticism scores from a total sample of 18 742 Australian twin individuals and their siblings. All participants completed detailed Composite International Diagnostic Interview from which diagnoses of Diagnostic and Statistical Manual of Mental Disorders (DSM)-IV depression and anxiety disorders were determined. Six hundred and seventy-four participants had a diagnosis of anxiety and/or depression from 492 families. The BDNF Val66Met and six DTNBP1 (rs3213207, rs1011313, rs2619528, rs760761, rs1018381, rs2619538) SNPs were genotyped on samples from study participants (n=2045 from 987 families) and, where possible, their parents (n=787). Family-based association tests were conducted between the individual SNPs and the DTNBP1 six SNP haplotypes and anxiety, depression, and neuroticism. RESULTS: We found no convincing evidence for association between any of the variants studied and anxiety, depression, or neuroticism. CONCLUSION: This study sample is relatively large but our results do not support an association between BDNF Val66Met and anxiety, depression, or neuroticisim. DTNBP1 haplotypes, which have been found to be risk factors for schizophrenia, are unlikely to be risk factors for anxiety and depression.


Asunto(s)
Ansiedad/genética , Factor Neurotrófico Derivado del Encéfalo/genética , Depresión/genética , Proteínas Asociadas a la Distrofina/genética , Neuropéptidos/genética , Trastornos Neuróticos/genética , Adulto , Frecuencia de los Genes , Humanos , Desequilibrio de Ligamiento , Polimorfismo de Nucleótido Simple , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
19.
Cancer Res ; 68(11): 4163-72, 2008 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-18519675

RESUMEN

We applied whole-genome single-nucleotide polymorphism arrays to define a comprehensive genetic profile of 23 esophageal adenocarcinoma (EAC) primary tumor biopsies based on loss of heterozygosity (LOH) and DNA copy number changes. Alterations were common, averaging 97 (range, 23-208) per tumor. LOH and gains averaged 33 (range, 3-83) and 31 (range, 11-73) per tumor, respectively. Copy neutral LOH events averaged 27 (range, 7-57) per EAC. We noted 126 homozygous deletions (HD) across the EAC panel (range, 0-11 in individual tumors). Frequent HDs within FHIT (17 of 23), WWOX (8 of 23), and DMD (6 of 23) suggest a role for common fragile sites or genomic instability in EAC etiology. HDs were also noted for known tumor suppressor genes (TSG), including CDKN2A, CDKN2B, SMAD4, and GALR1, and identified PDE4D and MGC48628 as potentially novel TSGs. All tumors showed LOH for most of chromosome 17p, suggesting that TSGs other than TP53 may be targeted. Frequent gains were noted around MYC (13 of 23), BCL9 (12 of 23), CTAGE1 (14 of 23), and ZNF217 (12 of 23). Thus, we have confirmed previous reports indicating frequent changes to FHIT, CDKN2A, TP53, and MYC in EAC and identified additional genes of interest. Meta-analysis of previous genome-wide EAC studies together with the data presented here highlighted consistent regions of gain on 8q, 18q, and 20q and multiple LOH regions on 4q, 5q, 17p, and 18q, suggesting that more than one gene may be targeted on each of these chromosome arms. The focal gains and deletions documented here are a step toward identifying the key genes involved in EAC development.


Asunto(s)
Adenocarcinoma/genética , Neoplasias Esofágicas/genética , Genoma , Polimorfismo de Nucleótido Simple , Mapeo Cromosómico , Perfilación de la Expresión Génica , Humanos , Análisis de Secuencia por Matrices de Oligonucleótidos
20.
Am J Med Genet B Neuropsychiatr Genet ; 147B(7): 1314-8, 2008 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-18384078

RESUMEN

The Val158Met polymorphism of the gene encoding catechol-O-methyltransferase (COMT) is one of the most widely tested variants for association with psychiatric disorders, but replication has been inconsistent including both sex limitation and heterogeneity of the associated allele. In this study we investigate the association between three SNPs from COMT and anxiety and depression disorders and neuroticism all measured within the same study sample. Participants were selected as sibling pairs (or multiples) that were either concordant or discordant for extreme neuroticism scores from a total sample of 18,742 Australian twin individuals and their siblings. All participants completed the Composite International Diagnostic Interview (CIDI) from which diagnoses of DSM-IV depression and anxiety disorders were determined. Of the participants, 674 had a diagnosis of anxiety and/or depression from 492 families. Study participants (n = 2,045 from 987 families) plus, where possible, their parents were genotyped for rs737865, rs4680 (Val158Met), and rs165599. Using family based tests we looked for association between these variants and neuroticism, depression, anxiety, panic disorder and agarophobia (PDAG) and obsessive compulsive disorder. We found no convincing evidence for association either in allelic or genotypic tests for the total sample or when the sample was stratified by sex. Haplotype T-G-G showed weak association (P = 0.042) with PDAG before correction for multiple testing; association between this haplotype and schizophrenia has been previously reported in an Australian sample.


Asunto(s)
Ansiedad/genética , Catecol O-Metiltransferasa/genética , Depresión/genética , Predisposición Genética a la Enfermedad/genética , Trastornos Neuróticos/genética , Australia , Salud de la Familia , Femenino , Genotipo , Humanos , Masculino , Polimorfismo de Nucleótido Simple , Factores Sexuales , Hermanos , Gemelos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...