Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
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
3.
Sci Rep ; 11(1): 20256, 2021 10 12.
Artigo em Inglês | MEDLINE | ID: mdl-34642383

RESUMO

Maintenance of genomic stability is critical to prevent diseases such as cancer. As such, eukaryotic cells have multiple pathways to efficiently detect, signal and repair DNA damage. One common form of exogenous DNA damage comes from ultraviolet B (UVB) radiation. UVB generates cyclobutane pyrimidine dimers (CPD) that must be rapidly detected and repaired to maintain the genetic code. The nucleotide excision repair (NER) pathway is the main repair system for this type of DNA damage. Here, we determined the role of the human Single-Stranded DNA Binding protein 2, hSSB2, in the response to UVB exposure. We demonstrate that hSSB2 levels increase in vitro and in vivo after UVB irradiation and that hSSB2 rapidly binds to chromatin. Depletion of hSSB2 results in significantly decreased Replication Protein A (RPA32) phosphorylation and impaired RPA32 localisation to the site of UV-induced DNA damage. Delayed recruitment of NER protein Xeroderma Pigmentosum group C (XPC) was also observed, leading to increased cellular sensitivity to UVB. Finally, hSSB2 was shown to have affinity for single-strand DNA containing a single CPD and for duplex DNA with a two-base mismatch mimicking a CPD moiety. Altogether our data demonstrate that hSSB2 is involved in the cellular response to UV exposure.


Assuntos
Reparo do DNA , Proteínas de Ligação a DNA/metabolismo , Proteína de Replicação A/metabolismo , Raios Ultravioleta/efeitos adversos , Animais , Linhagem Celular , Cromatina/metabolismo , Dano ao DNA , Proteínas de Ligação a DNA/genética , Regulação da Expressão Gênica/efeitos da radiação , Células HeLa , Humanos , Fosforilação/efeitos da radiação , Regulação para Cima
4.
Clin Cancer Res ; 27(9): 2624-2635, 2021 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-33589432

RESUMO

PURPOSE: G9a histone methyltransferase exerts oncogenic effects in several tumor types and its inhibition promotes anticancer effects. However, the impact on checkpoint inhibitor blockade response and the utility of G9a or its target genes as a biomarker is poorly studied. We aimed to examine whether G9a inhibition can augment the efficacy of checkpoint inhibitor blockade and whether LC3B, a G9a target gene, can predict treatment response. EXPERIMENTAL DESIGN: Clinical potential of LC3B as a biomarker of checkpoint inhibitor blockade was assessed using patient samples including tumor biopsies and circulating tumor cells from liquid biopsies. Efficacy of G9a inhibition to enhance checkpoint inhibitor blockade was examined using a mouse model. RESULTS: Patients with melanoma who responded to checkpoint inhibitor blockade were associated with not only a higher level of tumor LC3B but also a higher proportion of cells expressing LC3B. A higher expression of MAP1LC3B or LC3B protein was associated with longer survival and lower incidence of acquired resistance to checkpoint inhibitor blockade, suggesting LC3B as a potential predictive biomarker. We demonstrate that G9a histone methyltransferase inhibition is able to not only robustly induce LC3B level to augment the efficacy of checkpoint inhibitor blockade, but also induces melanoma cell death. CONCLUSIONS: Checkpoint inhibitor blockade response is limited to a subset of the patient population. These results have implications for the development of LC3B as a predictive biomarker of checkpoint inhibitor blockade to guide patient selection, as well as G9a inhibition as a strategy to extend the proportion of patients responding to immunotherapy.


Assuntos
Inibidores Enzimáticos/farmacologia , Histona-Lisina N-Metiltransferase/antagonistas & inibidores , Inibidores de Checkpoint Imunológico/farmacologia , Melanoma/metabolismo , Animais , Biomarcadores Tumorais , Linhagem Celular Tumoral , Modelos Animais de Doenças , Sinergismo Farmacológico , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Antígenos de Histocompatibilidade/genética , Antígenos de Histocompatibilidade/metabolismo , Histona-Lisina N-Metiltransferase/genética , Histona-Lisina N-Metiltransferase/metabolismo , Humanos , Inibidores de Checkpoint Imunológico/uso terapêutico , Melanoma/diagnóstico , Melanoma/tratamento farmacológico , Melanoma/genética , Camundongos , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Células Neoplásicas Circulantes , Prognóstico , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Sci Rep ; 11(1): 207, 2021 01 08.
Artigo em Inglês | MEDLINE | ID: mdl-33420238

RESUMO

The long-standing perception of Protein Kinase C (PKC) as a family of oncoproteins has increasingly been challenged by evidence that some PKC isoforms may act as tumor suppressors. To explore the hypothesis that activation, rather than inhibition, of these isoforms is critical for anticancer activity, we isolated and characterized a family of 16 novel phorboids closely-related to tigilanol tiglate (EBC-46), a PKC-activating epoxytigliane showing promising clinical safety and efficacy for intratumoral treatment of cancers. While alkyl branching features of the C12-ester influenced potency, the 6,7-epoxide structural motif and position was critical to PKC activation in vitro. A subset of the 6,7-epoxytiglianes were efficacious against established tumors in mice; which generally correlated with in vitro activation of PKC. Importantly, epoxytiglianes without evidence of PKC activation showed limited antitumor efficacy. Taken together, these findings provide a strong rationale to reassess the role of PKC isoforms in cancer, and suggest in some situations their activation can be a promising strategy for anticancer drug discovery.


Assuntos
Antineoplásicos/química , Antineoplásicos/farmacologia , Diterpenos/química , Diterpenos/farmacologia , Proteína Quinase C/metabolismo , Animais , Linhagem Celular Tumoral , Ativação Enzimática/efeitos dos fármacos , Camundongos , Transdução de Sinais/efeitos dos fármacos
6.
Chemistry ; 26(59): 13372-13377, 2020 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-32991008

RESUMO

The tigliane ring system, which encompasses iconic members such as phorbol and TPA, is widely renowned due to numerous observations of displaying potent biological activity, and subsequent use as mainstream biochemical tools. Traditionally, naturally occurring phorboids are regarded as tumor promotors through PKC activation, although in recent times more highly oxidized natural derivatives have been identified as anti-tumor agents. In the view that only limited synthetic investigations toward skeletal stereochemical modification have been undertaken, non-natural systems could be useful for a better understanding of the tigliane pharmacophore via interrogation of cellular sensitivity. In this context the concise construction of a number of highly functionalized non-natural D-ring inverted phorbol esters were synthesized, via a rhodium-catalyzed [4+3] cycloaddition, and biologically evaluated using a range of cancer cell lines. The biological results highlight the notion that subtle changes in structure have dramatic effects on potency. Furthermore, although the non-natural derivatives did not outcompete the natural systems in the PKC-activation sensitive MCF7 cancer cell line, they outperformed in other cancer cell lines (MM96L and CAL27). This observation strongly suggested an alternate mode of action not involving activation of PKC, but instead involves thiol addition as indicated by glutathione addition and NF-κB reporter activity.


Assuntos
Neoplasias , Forbóis , Proteína Quinase C/química , Compostos de Sulfidrila/química , Linhagem Celular , Humanos
8.
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
9.
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
10.
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
11.
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.
Methods Mol Biol ; 1267: 251-61, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25636472

RESUMO

Mouse models of melanoma have proven invaluable in the delineation of key molecular events involved in disease progression in humans and provide potential preclinical models for therapeutic testing (Damsky and Bosenberg, Pigment Cell Melanoma Res 25(4):404-405, 2012; Walker et al., Pigment Cell Melanoma Res 24(6):1158-1176, 2011). Here we concentrate on the clinicopathological analysis of melanocytic tumors.


Assuntos
Modelos Animais de Doenças , Melanoma/patologia , Animais , Dermoscopia , Amarelo de Eosina-(YS)/metabolismo , Hematoxilina/metabolismo , Imuno-Histoquímica , Melanócitos/patologia , Melanócitos/efeitos da radiação , Melanoma/diagnóstico , Melanoma/etiologia , Melanoma/metabolismo , Glicoproteínas de Membrana/metabolismo , Camundongos , Oxirredutases/metabolismo , Inclusão em Parafina , Fatores de Transcrição SOXE/metabolismo , Neoplasias Cutâneas/diagnóstico , Neoplasias Cutâneas/etiologia , Neoplasias Cutâneas/metabolismo , Neoplasias Cutâneas/patologia , Coloração e Rotulagem , Fixação de Tecidos , Raios Ultravioleta/efeitos adversos
14.
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
16.
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
17.
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
18.
Exp Dermatol ; 21(9): 676-81, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22742762

RESUMO

It has been shown that gene mutations which drive the development of malignant melanoma (MM) in humans also lead to emergence of MM when engineered mice. However, little attention has been paid to the clinical and histopathological features of melanocytic lesions and their natural history in a given mouse model. This knowledge is crucial to enable us to understand how engineered mutations influence the initiation and evolution of melanocytic lesions, and/or for the use of mice as a preclinical model to test specific treatments. We recently reported the development of melanocytic proliferations along the spectrum of naevi to MM in a Cdk4 ( R24C/R24C ) ::Tyr- NRAS ( Q ) ( 61K ) mouse model. In this study, we followed the development of lesions over time using digital photography and dermoscopy with the aim to correlate the clinical and histopathological features of lesions developing in this model. We identified two types of lesions. The first are slow-growing dermal MMs that emanate from dermal naevi. The second did not emanate from naevi, grew rapidly, and appeared to be solely confined to the subcutaneous fat. We present a simple staging system for the MMs that progress from naevi, based on depth of extension into the dermis and subcutis. This represents a blueprint for documentation and follow-up of MMs in the live animal, which is critical for the proper use of murine melanoma models.


Assuntos
Modelos Animais de Doenças , Melanoma/patologia , Nevo/patologia , Neoplasias Cutâneas/patologia , Animais , Quinase 4 Dependente de Ciclina/genética , Dermoscopia , Genes ras , Engenharia Genética , Imuno-Histoquímica , Melanoma/genética , Camundongos , Nevo/genética , Fotografação , Neoplasias Cutâneas/genética
20.
J Invest Dermatol ; 132(4): 1263-71, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22189786

RESUMO

Skin cancer is the most prevalent cancer worldwide and is primarily caused by chronic UV exposure. Here, we describe the topical field-directed treatment of SKH1/hr mice with UVB-damaged skin with ingenol mebutate, a new topical drug shown to be effective for the treatment of actinic keratosis (AK). Application of 0.05% ingenol mebutate gel to photo-damaged skin resulted in a ≈70% reduction in the number of skin lesions that subsequently emerged compared with placebo treatment. Ingenol mebutate treatment also reduced the number of mutant p53 keratinocyte patches by ≈70%. The treatment resulted in epidermal cell death, acute inflammation, recruitment of neutrophils, hemorrhage, and eschar formation, all of which resolved over several weeks. Ingenol mebutate field-directed treatment might thus find utility in the removal of subclinical precancerous cells from UV-damaged skin. Field-directed treatment may be particularly suitable for patients who have AKs surrounded by UV-damaged skin.


Assuntos
Diterpenos/uso terapêutico , Queratinócitos/metabolismo , Queratinócitos/patologia , Ceratose Actínica/tratamento farmacológico , Lesões Pré-Cancerosas/tratamento farmacológico , Pele/efeitos da radiação , Proteína Supressora de Tumor p53/metabolismo , Raios Ultravioleta/efeitos adversos , Animais , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Apoptose/efeitos dos fármacos , Carcinoma de Células Escamosas/prevenção & controle , Modelos Animais de Doenças , Diterpenos/farmacologia , Relação Dose-Resposta a Droga , Queratinócitos/efeitos dos fármacos , Ceratose Actínica/patologia , Masculino , Camundongos , Camundongos Pelados , Mutação/genética , Neoplasias Induzidas por Radiação/prevenção & controle , Lesões Pré-Cancerosas/patologia , Pele/efeitos dos fármacos , Pele/patologia , Neoplasias Cutâneas/prevenção & controle , Proteína Supressora de Tumor p53/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...