Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
J Immunother Cancer ; 12(4)2024 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-38658031

RESUMO

BACKGROUND: Tigilanol tiglate (TT) is a protein kinase C (PKC)/C1 domain activator currently being developed as an intralesional agent for the treatment of various (sub)cutaneous malignancies. Previous work has shown that intratumoral (I.T.) injection of TT causes vascular disruption with concomitant tumor ablation in several preclinical models of cancer, in addition to various (sub)cutaneous tumors presenting in the veterinary clinic. TT has completed Phase I dose escalation trials, with some patients showing signs of abscopal effects. However, the exact molecular details underpinning its mechanism of action (MoA), together with its immunotherapeutic potential in oncology remain unclear. METHODS: A combination of microscopy, luciferase assays, immunofluorescence, immunoblotting, subcellular fractionation, intracellular ATP assays, phagocytosis assays and mixed lymphocyte reactions were used to probe the MoA of TT in vitro. In vivo studies with TT used MM649 xenograft, CT-26 and immune checkpoint inhibitor refractory B16-F10-OVA tumor bearing mice, the latter with or without anti-programmed cell death 1 (PD-1)/anti-cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) mAb treatment. The effect of TT at injected and non-injected tumors was also assessed. RESULTS: Here, we show that TT induces the death of endothelial and cancer cells at therapeutically relevant concentrations via a caspase/gasdermin E-dependent pyroptopic pathway. At therapeutic doses, our data demonstrate that TT acts as a lipotoxin, binding to and promoting mitochondrial/endoplasmic reticulum (ER) dysfunction (leading to unfolded protein responsemt/ER upregulation) with subsequent ATP depletion, organelle swelling, caspase activation, gasdermin E cleavage and induction of terminal necrosis. Consistent with binding to ER membranes, we found that TT treatment promoted activation of the integrated stress response together with the release/externalization of damage-associated molecular patterns (HMGB1, ATP, calreticulin) from cancer cells in vitro and in vivo, characteristics indicative of immunogenic cell death (ICD). Confirmation of ICD in vivo was obtained through vaccination and rechallenge experiments using CT-26 colon carcinoma tumor bearing mice. Furthermore, TT also reduced tumor volume, induced immune cell infiltration, as well as improved survival in B16-F10-OVA tumor bearing mice when combined with immune checkpoint blockade. CONCLUSIONS: These data demonstrate that TT is an oncolytic small molecule with multiple targets and confirms that cell death induced by this compound has the potential to augment antitumor responses to immunotherapy.


Assuntos
Inibidores de Checkpoint Imunológico , Morte Celular Imunogênica , Animais , Camundongos , Morte Celular Imunogênica/efeitos dos fármacos , Humanos , Inibidores de Checkpoint Imunológico/farmacologia , Inibidores de Checkpoint Imunológico/uso terapêutico , Linhagem Celular Tumoral , Feminino , Ensaios Antitumorais Modelo de Xenoenxerto , Neoplasias/tratamento farmacológico , Neoplasias/imunologia , Neoplasias/terapia
2.
J Nat Prod ; 85(8): 1959-1966, 2022 08 26.
Artigo em Inglês | MEDLINE | ID: mdl-35973043

RESUMO

The kernels of the Australian blushwood tree (Fontainea picrosperma) are the source of the veterinary anticancer drug tigilanol tiglate (2a, Stelfonta) and contain a concentration of phorboids significantly higher than croton oil, the only abundant source of these compounds previously known. The oily matrix of the blushwood kernels is composed of free fatty acids and not by glycerides as found in croton oil. By active partitioning, it was therefore possible to recover and characterize for the first time a cryptic tigliane fraction, that is, the diterpenoid fraction that, because of its lipophilicity, could not be obtained by solvent partition of crude extracts. The cryptic tigliane fraction accounted for ca. 30% of the tigliane kernel titer and was quantified by 1H NMR spectroscopy and profiled by HPLC-MS. Long-chain (linoleates and/or oleates) 20-acyl derivatives of the epoxytigliane diesters tigilanol tiglate (EBC-46, 2a), EBC-47 (4a), EBC-59 (5a), EBC-83 (6a), and EBC-177 (7a) were identified. By chemoselective acylation of EBC-46 (2a) and EBC-177 (7a) the natural triesters 2b and 7b and a selection of analogues were prepared to assist identification of the natural compounds. The presence of a free C-20 hydroxy group is a critical requirement for PKC activation by phorbol esters. The unexpected activity of 20-linoleoyl triester 2b in a cytotoxicity assay based on PKC activation was found to be related mainly to its hydrolysis to tigilanol tiglate (2a) under the prolonged conditions of the assay, while other esters were inactive. Significant differences between the esterification profile of the epoxytigliane di- and triesters exist in F. picrosperma, suggesting a precise, yet elusive, blueprint of acyl decoration for the tigliane polyol 5-hydroxyepoxyphorbol.


Assuntos
Euphorbiaceae , Forbóis , Austrália , Óleo de Cróton , Árvores
4.
Am J Respir Crit Care Med ; 205(3): 300-312, 2022 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-34860143

RESUMO

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.


Assuntos
Asma/imunologia , Proteína HMGB1/metabolismo , Interleucina-33/metabolismo , Receptores Purinérgicos P2/metabolismo , Animais , Asma/metabolismo , Asma/fisiopatologia , Biomarcadores/metabolismo , Estudos de Casos e Controles , Progressão da Doença , Ensaio de Imunoadsorção Enzimática , Células Epiteliais/metabolismo , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos C57BL
5.
Exp Dermatol ; 29(5): 450-461, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32145039

RESUMO

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.


Assuntos
Proteínas Morfogenéticas Ósseas/metabolismo , Mapeamento Cromossômico , Estudo de Associação Genômica Ampla , Cabelo/fisiologia , Polimorfismo Genético , Transdução de Sinais , Proteínas Wnt/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Cruzamentos Genéticos , Derme/metabolismo , Ligação Genética , Camundongos , Fenótipo , Locos de Características Quantitativas , Especificidade da Espécie , Proteína 1 Relacionada a Twist/genética
7.
Elife ; 82019 01 25.
Artigo em Inglês | MEDLINE | ID: mdl-30681412

RESUMO

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.


Assuntos
Melanoma/genética , Neoplasias Cutâneas/genética , Raios Ultravioleta , Animais , Animais Recém-Nascidos , Animais não Endogâmicos , Proliferação de Células , Mapeamento Cromossômico , Cromossomos de Mamíferos/genética , Modelos Animais de Doenças , Regulação Neoplásica da Expressão Gênica , Redes Reguladoras de Genes , Loci Gênicos , Humanos , Melanócitos/metabolismo , Melanócitos/patologia , Melanoma/patologia , Camundongos Transgênicos , Proteínas Monoméricas de Ligação ao GTP/genética , Proteínas Proto-Oncogênicas B-raf/genética , Locos de Características Quantitativas/genética , Reprodutibilidade dos Testes , Pele/metabolismo , Pele/patologia , Neoplasias Cutâneas/patologia , Melanoma Maligno Cutâneo
8.
J Invest Dermatol ; 139(1): 177-185, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30009829

RESUMO

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.


Assuntos
Citocinas/genética , Regulação Neoplásica da Expressão Gênica , Queratinócitos/metabolismo , Nevo Pigmentado/genética , RNA Neoplásico/genética , Neoplasias Cutâneas/genética , Animais , Citocinas/biossíntese , Estudo de Associação Genômica Ampla , Humanos , Queratinócitos/patologia , Camundongos , Camundongos Knockout , Nevo Pigmentado/metabolismo , Nevo Pigmentado/patologia , Neoplasias Cutâneas/metabolismo , Neoplasias Cutâneas/patologia
9.
J Invest Dermatol ; 138(4): 893-902, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29138054

RESUMO

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.


Assuntos
Endotelina-1/metabolismo , Regulação Neoplásica da Expressão Gênica , Proteínas Hedgehog/genética , Queratinócitos/metabolismo , Neoplasias Experimentais , Nevo Pigmentado/genética , Neoplasias Cutâneas/genética , Regulação para Cima , Animais , Feminino , Proteínas Hedgehog/biossíntese , Humanos , Queratinócitos/patologia , Masculino , Melanócitos/metabolismo , Melanócitos/patologia , Camundongos , Camundongos Knockout , Nevo Pigmentado/metabolismo , Nevo Pigmentado/patologia , Transdução de Sinais , Neoplasias Cutâneas/metabolismo , Neoplasias Cutâneas/patologia , Ativação Transcricional , Células Tumorais Cultivadas
10.
Pigment Cell Melanoma Res ; 29(4): 459-64, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27155367

RESUMO

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.


Assuntos
Moléculas de Adesão Celular/genética , GTP Fosfo-Hidrolases/genética , Melanócitos/patologia , Proteínas de Membrana/genética , Mutação , Nevo/congênito , Neoplasias Cutâneas/congênito , Animais , Células Cultivadas , Derme/metabolismo , Derme/patologia , Feminino , Folículo Piloso/metabolismo , Folículo Piloso/patologia , Humanos , Queratinócitos/metabolismo , Queratinócitos/patologia , Masculino , Melanócitos/metabolismo , Camundongos , Camundongos Knockout , Nevo/patologia , Neoplasias Cutâneas/patologia
12.
Photochem Photobiol Sci ; 14(6): 1179-89, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25966309

RESUMO

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.


Assuntos
Folículo Piloso/citologia , Folículo Piloso/efeitos da radiação , Melanócitos/efeitos da radiação , Raios Ultravioleta , Animais , Proliferação de Células/efeitos da radiação , Dano ao DNA , Imuno-Histoquímica , Melanócitos/citologia , Camundongos , Pele/citologia , Pele/efeitos da radiação , Células-Tronco/citologia , Células-Tronco/efeitos da radiação
13.
Curr Probl Dermatol ; 46: 42-8, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25561205

RESUMO

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.


Assuntos
Carcinoma de Células Escamosas , Modelos Animais de Doenças , Ceratose Actínica , Neoplasias Experimentais , Neoplasias Cutâneas , Animais , Carcinoma de Células Escamosas/induzido quimicamente , Carcinoma de Células Escamosas/genética , Ceratose Actínica/induzido quimicamente , Ceratose Actínica/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neoplasias Experimentais/induzido quimicamente , Neoplasias Experimentais/genética , Neoplasias Cutâneas/induzido quimicamente , Neoplasias Cutâneas/genética
14.
J Invest Dermatol ; 134(7): 1991-1997, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24509534

RESUMO

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.


Assuntos
Macrófagos/patologia , Melanoma/patologia , Neoplasias Induzidas por Radiação/patologia , Neutrófilos/patologia , Neoplasias Cutâneas/patologia , Raios Ultravioleta/efeitos adversos , Fatores Etários , Animais , Animais Recém-Nascidos , Proliferação de Células/efeitos da radiação , Citometria de Fluxo , Macrófagos/efeitos da radiação , Melanoma/epidemiologia , Melanoma/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Células Mieloides/patologia , Células Mieloides/efeitos da radiação , Neoplasias Induzidas por Radiação/epidemiologia , Neoplasias Induzidas por Radiação/imunologia , Neutrófilos/efeitos da radiação , Fatores de Risco , Neoplasias Cutâneas/epidemiologia , Neoplasias Cutâneas/imunologia
15.
Pigment Cell Melanoma Res ; 26(5): 731-4, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23724991

RESUMO

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.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Transdiferenciação Celular , Quinase 4 Dependente de Ciclina/metabolismo , Melanoma/metabolismo , Melanoma/patologia , Neurônios/patologia , Proteólise , Proteína Supressora de Tumor p53/metabolismo , Animais , Transdiferenciação Celular/genética , Quinase 4 Dependente de Ciclina/genética , Regulação para Baixo , Regulação Neoplásica da Expressão Gênica , Humanos , Melanoma/genética , Camundongos , Neurônios/metabolismo , Regulação para Cima/genética
16.
J Invest Dermatol ; 133(7): 1803-12, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23321920

RESUMO

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.


Assuntos
Antígenos Ly/metabolismo , Proliferação de Células/efeitos da radiação , Antígenos de Histocompatibilidade Classe II/metabolismo , Macrófagos/imunologia , Melanócitos/metabolismo , Receptores CCR2/metabolismo , Raios Ultravioleta , Animais , Animais Recém-Nascidos , Transformação Celular Neoplásica/patologia , Interleucina-17/metabolismo , Macrófagos/patologia , Melanócitos/patologia , Melanócitos/efeitos da radiação , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenótipo , Receptores CCR2/deficiência , Receptores CCR2/genética , Fatores de Risco , Pele/metabolismo , Pele/patologia , Queimadura Solar/complicações , Fatores de Tempo
19.
Pigment Cell Melanoma Res ; 23(6): 771-80, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20718941

RESUMO

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.


Assuntos
Melanoma/metabolismo , Nevo/metabolismo , Lesões Pré-Cancerosas/patologia , Proteína do Retinoblastoma/metabolismo , Transdução de Sinais , Proteína Supressora de Tumor p14ARF/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Animais , Animais Recém-Nascidos , Contagem de Células , Movimento Celular/efeitos da radiação , Proliferação de Células/efeitos da radiação , Quinase 4 Dependente de Ciclina/metabolismo , Melanócitos/patologia , Melanócitos/efeitos da radiação , Melanoma/genética , Melanoma/patologia , Camundongos , Modelos Biológicos , Nevo/genética , Nevo/patologia , Penetrância , Lesões Pré-Cancerosas/metabolismo , Transdução de Sinais/efeitos da radiação , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/metabolismo , Neoplasias Cutâneas/patologia , Raios Ultravioleta
20.
J Alzheimers Dis ; 20(2): 561-7, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20182055

RESUMO

Alzheimer's disease (AD) patients commonly suffer from behavioral and psychological symptoms of dementia (BPSD). Variants within the neuregulin-1 (NRG1) gene have been investigated both in early onset psychiatric disorders, such as schizophrenia and recently in AD patients with psychosis. In this study, we analyzed NRG1 variants in AD patients with and without psychosis. Our large cohort of 399 probable AD patients had longitudinal information on the BPSD, which was used to dichotomize patients into whether they had ever suffered from psychotic symptoms within the study period. The NRG1 single nucleotide polymorphisms rs3924999, rs35753505 (SNP8NRG221533) and the microsatellites 478B14-848 and 420M9-1395 were investigated for association with psychosis using genotype, allele, and haplotype analyses. No associations were found between any of these variants or haplotypic combinations with delusions, hallucinations, psychosis, or elation/mania in our cohort. Positive associations with polymorphisms and haplotype combinations of NRG1 have been reported in psychiatric disorders. One previous study found an association with psychosis in AD, with a SNP outside the haplotype block first reported for association with schizophrenia. We found no association with any of these variants in our cohort. Further investigations of this region on chromosome 8 are clearly required, with replication in different large longitudinal cohorts.


Assuntos
Doença de Alzheimer/complicações , Neuregulina-1/genética , Transtornos Psicóticos/etiologia , Transtornos Psicóticos/genética , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/genética , Estudos de Coortes , Feminino , Frequência do Gene , Estudo de Associação Genômica Ampla , Genótipo , Humanos , Modelos Logísticos , Masculino , Repetições de Microssatélites/genética , Polimorfismo de Nucleotídeo Único/genética , Escalas de Graduação Psiquiátrica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA