Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 14(1): 1328, 2023 03 10.
Artigo em Inglês | MEDLINE | ID: mdl-36899004

RESUMO

The TINCR (Terminal differentiation-Induced Non-Coding RNA) gene is selectively expressed in epithelium tissues and is involved in the control of human epidermal differentiation and wound healing. Despite its initial report as a long non-coding RNA, the TINCR locus codes for a highly conserved ubiquitin-like microprotein associated with keratinocyte differentiation. Here we report the identification of TINCR as a tumor suppressor in squamous cell carcinoma (SCC). TINCR is upregulated by UV-induced DNA damage in a TP53-dependent manner in human keratinocytes. Decreased TINCR protein expression is prevalently found in skin and head and neck squamous cell tumors and TINCR expression suppresses the growth of SCC cells in vitro and in vivo. Consistently, Tincr knockout mice show accelerated tumor development following UVB skin carcinogenesis and increased penetrance of invasive SCCs. Finally, genetic analyses identify loss-of-function mutations and deletions encompassing the TINCR gene in SCC clinical samples supporting a tumor suppressor role in human cancer. Altogether, these results demonstrate a role for TINCR as protein coding tumor suppressor gene recurrently lost in squamous cell carcinomas.


Assuntos
Carcinoma de Células Escamosas , Neoplasias de Cabeça e Pescoço , RNA Longo não Codificante , Animais , Camundongos , Humanos , Ubiquitina/metabolismo , Carcinoma de Células Escamosas/genética , Genes Supressores de Tumor , Queratinócitos/metabolismo , Neoplasias de Cabeça e Pescoço/genética , RNA Longo não Codificante/genética , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Micropeptídeos
4.
Sci Rep ; 9(1): 13072, 2019 09 10.
Artigo em Inglês | MEDLINE | ID: mdl-31506465

RESUMO

The study of dominantly heritable cancers has provided insights about tumor development. Gorlin syndrome (GS) is an autosomal dominant disorder wherein affected individuals develop multiple basal cell carcinomas (BCCs) of the skin. We developed a murine model of Ptch1 haploinsufficiency on an ornithine decarboxylase (ODC) transgenic background (Ptch1+/-/ODCt/C57BL/6) that is more sensitive to BCCs growth as compared with Ptch1+/+/ODCt/C57BL/6 littermates. Ptch1+/-/ODCt/C57BL/6 mice show an altered metabolic landscape in the phenotypically normal skin, including restricted glucose availability, restricted ribose/deoxyribose flow and NADPH production, an accumulation of α-ketoglutarate, aconitate, and citrate that is associated with reversal of the tricarboxylic acid cycle, coupled with increased ketogenic/lipogenic activity via acetyl-CoA, 3-hydroybutyrate, and cholesterol metabolites. Also apparent was an increased content/acetylation of amino-acids, glutamine and glutamate, in particular. Accordingly, metabolic alterations due to a single copy loss of Ptch1 in Ptch1+/-/ODCt/C57BL/6 heterozygous mice may provide insights about the cancer prone phenotype of BCCs in GS patients, including biomarkers/targets for early intervention.


Assuntos
Metabolismo Energético/genética , Haploinsuficiência , Ornitina Descarboxilase/genética , Receptor Patched-1/genética , Pele/metabolismo , Animais , Biologia Computacional/métodos , Perfilação da Expressão Gênica , Heterozigoto , Metabolismo dos Lipídeos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Via de Pentose Fosfato , Fenótipo , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/metabolismo , Neoplasias Cutâneas/patologia , Transcriptoma
5.
J Invest Dermatol ; 138(8): 1716-1725, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29550418

RESUMO

Currently available smoothened targeted therapies in patients with basal cell nevus syndrome are associated with substantial tumor recurrence and clinical resistance. Strategies bypassing smoothened and/or identifying additional downstream components of the Hedgehog pathway could provide novel antitumor targets with a better therapeutic index. Sry-related high mobility group box 9 (SOX9) is a Hedgehog/glioma-associated oncogene homolog-regulated transcription factor known to be overexpressed in basal cell carcinomas (BCCs). A sequence motif search for SOX9-responsive elements identified three motifs in the promoter region of mammalian target of rapamycin (mTOR). In murine BCC cells, SOX9 occupies the mTOR promoter and induces its transcriptional activity. Short hairpin RNA (shRNA)-mediated knockdown of SOX9, as well as smoothened inhibition by itraconazole and vismodegib, reduces mTOR expression and the phosphorylation of known downstream mTOR targets. These effects culminate in diminishing the proliferative capacity of BCC cells, demonstrating a direct mechanistic link between the Hedgehog and mTOR pathways capable of driving BCC growth. Furthermore, rapamycin, a pharmacologic mTOR inhibitor, suppressed the growth of UV-induced BCCs in Ptch1+/-/SKH-1 mice, a model that closely mimics the accelerated BCC growth pattern of patients with basal cell nevus syndrome. Our data demonstrate that Hedgehog signaling converges on mTOR via SOX9, and highlight the SOX9-mTOR axis as a viable additional target downstream of smoothened that could enhance tumor elimination in patients with BCC.


Assuntos
Carcinoma Basocelular/genética , Carcinoma de Células Escamosas/genética , Fatores de Transcrição SOX9/metabolismo , Neoplasias Cutâneas/genética , Serina-Treonina Quinases TOR/genética , Animais , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Carcinoma Basocelular/tratamento farmacológico , Carcinoma Basocelular/patologia , Carcinoma de Células Escamosas/tratamento farmacológico , Carcinoma de Células Escamosas/patologia , Linhagem Celular Tumoral , Proliferação de Células/genética , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes , Células HEK293 , Proteínas Hedgehog/metabolismo , Humanos , Camundongos , Camundongos Pelados , Recidiva Local de Neoplasia , Neoplasias Experimentais/tratamento farmacológico , Neoplasias Experimentais/etiologia , Neoplasias Experimentais/patologia , Fosforilação/efeitos dos fármacos , Regiões Promotoras Genéticas/genética , RNA Interferente Pequeno/metabolismo , Fatores de Transcrição SOX9/genética , Transdução de Sinais/genética , Pele/patologia , Neoplasias Cutâneas/tratamento farmacológico , Neoplasias Cutâneas/patologia , Receptor Smoothened/antagonistas & inibidores , Receptor Smoothened/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Análise Serial de Tecidos , Raios Ultravioleta/efeitos adversos
7.
PLoS One ; 11(9): e0163054, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27643989

RESUMO

Exposure to ultraviolet radiation (UVR) is a major risk factor for both melanoma and non-melanoma skin cancers. In addition to its mutagenic effect, UVR can also induce substantial transcriptional instability in skin cells affecting thousands of genes, including many cancer genes, suggesting that transcriptional instability may be another important etiological factor in skin photocarcinogenesis. In this study, we performed detailed transcriptomic profiling studies to characterize the kinetic changes in global gene expression in human keratinocytes exposed to different UVR conditions. We identified a subset of UV-responsive genes as UV signature genes (UVSGs) based on 1) conserved UV-responsiveness of this subset of genes among different keratinocyte lines; and 2) UV-induced persistent changes in their mRNA levels long after exposure. Interestingly, 11 of the UVSGs were shown to be critical to skin cancer cell proliferation and survival. Through computational Gene Set Enrichment Analysis, we demonstrated that a significant portion of the UVSGs were dysregulated in human skin squamous cell carcinomas, but not in other human malignancies. This highlights the potential and specificity of the UVSGs in clinical diagnosis of UV damage and stratification of skin cancer risk.


Assuntos
Neoplasias Cutâneas/genética , Transcriptoma , Raios Ultravioleta , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patologia , Proliferação de Células , Relação Dose-Resposta à Radiação , Perfilação da Expressão Gênica , Humanos , Neoplasias Cutâneas/patologia
8.
Cancer Prev Res (Phila) ; 9(10): 794-802, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27388747

RESUMO

Patients with basal cell nevus syndrome (BCNS), also known as Gorlin syndrome, develop numerous basal cell carcinomas (BCC) due to germline mutations in the tumor suppressor PTCH1 and aberrant activation of Hedgehog (Hh) signaling. Therapies targeted at components of the Hh pathway, including the smoothened (SMO) inhibitor vismodegib, can ablate these tumors clinically, but tumors recur upon drug discontinuation. Using SKH1-Ptch1+/- as a model that closely mimics the spontaneous and accelerated growth pattern of BCCs in patients with BCNS, we show that AKT1, a serine/threonine protein kinase, is intrinsically activated in keratinocytes derived from the skin of newborn Ptch1+/- mice in the absence of carcinogenic stimuli. Introducing Akt1 haplodeficiency in Ptch1+/- mice (Akt1+/- Ptch1+/-) significantly abrogated BCC growth. Similarly, pharmacological inhibition of AKT with perifosine, an alkyl phospholipid AKT inhibitor, diminished the growth of spontaneous and UV-induced BCCs. Our data demonstrate an obligatory role for AKT1 in BCC growth, and targeting AKT may help reduce BCC tumor burden in BCNS patients. Cancer Prev Res; 9(10); 794-802. ©2016 AACR.


Assuntos
Síndrome do Nevo Basocelular/patologia , Carcinoma Basocelular/patologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Neoplasias Cutâneas/patologia , Animais , Síndrome do Nevo Basocelular/metabolismo , Carcinoma Basocelular/metabolismo , Linhagem Celular , Modelos Animais de Doenças , Humanos , Queratinócitos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neoplasias Cutâneas/metabolismo
9.
Exp Dermatol ; 25(9): 708-13, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27119462

RESUMO

Solar ultraviolet radiation (UVR) is the major risk factor for skin carcinogenesis. To gain new insights into the molecular pathways mediating UVR effects in the skin, we performed comprehensive transcriptomic analyses to identify shared and distinctive molecular responses to UVR between human keratinocytes and melanocytes. Keratinocytes and melanocytes were irradiated with varying doses of UVB (10, 20 and 30 mJ/cm(2) ) then analysed by RNA-Seq at different time points post-UVB radiation (4, 24 and 72 h). Under basal conditions, keratinocytes and melanocytes expressed similar number of genes, although they each expressed a distinctive subset of genes pertaining to their specific cellular identity. Upon UVB radiation, keratinocytes displayed a clear pattern of time- and dose-dependent changes in gene expression that was different from melanocytes. The early UVB-responsive gene set (4 h post-UVR) differed significantly from delayed UVB-responsive gene sets (24 and 72 h). We also identified multiple novel UVB signature genes including PRSS23, SERPINH1, LCE3D and CNFN, which were conserved between melanocyte and keratinocyte lines from different individuals. Taken together, our findings elucidated both common and distinctive molecular features between melanocytes and keratinocytes and uncovered novel UVB signature genes that might be utilized to predict UVB photobiological effects on the skin.


Assuntos
Queratinócitos/efeitos da radiação , Melanócitos/efeitos da radiação , Sobrevivência Celular , Humanos , Queratinócitos/metabolismo , Melanócitos/metabolismo , Cultura Primária de Células , Transcriptoma , Raios Ultravioleta
10.
J Immunol ; 196(8): 3233-44, 2016 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-26944931

RESUMO

Atopic dermatitis, a chronic inflammatory skin disease with increasing prevalence, is closely associated with skin barrier defects. A cytokine related to disease severity and inhibition of keratinocyte differentiation is IL-31. To identify its molecular targets, IL-31-dependent gene expression was determined in three-dimensional organotypic skin models. IL-31-regulated genes are involved in the formation of an intact physical skin barrier. Many of these genes were poorly induced during differentiation as a consequence of IL-31 treatment, resulting in increased penetrability to allergens and irritants. Furthermore, studies employing cell-sorted skin equivalents in SCID/NOD mice demonstrated enhanced transepidermal water loss following s.c. administration of IL-31. We identified the IL-1 cytokine network as a downstream effector of IL-31 signaling. Anakinra, an IL-1R antagonist, blocked the IL-31 effects on skin differentiation. In addition to the effects on the physical barrier, IL-31 stimulated the expression of antimicrobial peptides, thereby inhibiting bacterial growth on the three-dimensional organotypic skin models. This was evident already at low doses of IL-31, insufficient to interfere with the physical barrier. Together, these findings demonstrate that IL-31 affects keratinocyte differentiation in multiple ways and that the IL-1 cytokine network is a major downstream effector of IL-31 signaling in deregulating the physical skin barrier. Moreover, by interfering with IL-31, a currently evaluated drug target, we will have to consider that low doses of IL-31 promote the antimicrobial barrier, and thus a complete inhibition of IL-31 signaling may be undesirable.


Assuntos
Dermatite Atópica/patologia , Interleucina-1alfa/metabolismo , Interleucina-1beta/metabolismo , Interleucinas/metabolismo , Junções Íntimas/metabolismo , Animais , Peptídeos Catiônicos Antimicrobianos/biossíntese , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Linhagem Celular , Proteínas Filagrinas , Humanos , Proteína Antagonista do Receptor de Interleucina 1/farmacologia , Interleucinas/farmacologia , Proteínas de Filamentos Intermediários/metabolismo , Queratinócitos/citologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Receptores de Interleucina-1/antagonistas & inibidores , Transdução de Sinais/fisiologia , Pele/citologia , Pele/crescimento & desenvolvimento , Junções Íntimas/efeitos dos fármacos
11.
Oncotarget ; 6(34): 36789-814, 2015 Nov 03.
Artigo em Inglês | MEDLINE | ID: mdl-26413810

RESUMO

Nevoid basal cell carcinoma syndrome (NBCCS) is a rare autosomal dominant disorder that is due, in large measure, to aberrant Shh signaling driven by mutations in the tumor suppressor gene Ptch1. Here, we describe the development of Ptch1+/-/ SKH-1 mice as a novel model of this disease. These animals manifest many features of NBCCS, including developmental anomalies and are remarkably sensitive to both ultraviolet (UVB) and ionizing radiation that drive the development of multiple BCCs. Just as in patients with NBCCS, Ptch1+/-/SKH-1 also spontaneously develops BCCs and other neoplasms such as rhabdomyomas/rhabdomyosarcomas. Administration of smoothened inhibitors (vismodegib/itraconazole/cyclopamine) or non-steroidal anti-inflammatory drug (sulindac/sulfasalazine) each result in partial resolution of BCCs in these animals. However, combined administration of these agents inhibits the growth of UVB-induced BCCs by >90%. Employing small molecule- and decoy-peptide-based approaches we further affirm that complete remission of BCCs could only be achieved by combined inhibition of p50-NFκB/Bcl3 and Shh signaling. We posit that Ptch1+/-/SKH-1 mice are a novel and relevant animal model for NBCCS. Understanding mechanisms that govern genetic predisposition to BCCs should facilitate our ability to identify and treat NBCCS gene carriers, including those at risk for sporadic BCCs while accelerating development of novel therapeutic modalities for these patients.


Assuntos
Síndrome do Nevo Basocelular/metabolismo , Proteínas Hedgehog/metabolismo , Subunidade p50 de NF-kappa B/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Neoplasias Cutâneas/metabolismo , Fatores de Transcrição/metabolismo , Animais , Proteína 3 do Linfoma de Células B , Síndrome do Nevo Basocelular/genética , Síndrome do Nevo Basocelular/patologia , Modelos Animais de Doenças , Feminino , Proteínas Hedgehog/genética , Humanos , Masculino , Camundongos , Camundongos Pelados , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Subunidade p50 de NF-kappa B/genética , Proteínas Proto-Oncogênicas/genética , Transdução de Sinais , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/patologia , Fatores de Transcrição/genética
12.
Cancer Res ; 74(18): 4967-75, 2014 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-25172843

RESUMO

The hedgehog (Hh) signaling pathway is considered to be a major signal transduction pathway during embryonic development, but it usually shuts down after birth. Aberrant Sonic hedgehog (Shh) activation during adulthood leads to neoplastic growth. Basal cell carcinoma (BCC) of the skin is driven by this pathway. Here, we summarize information related to the pathogenesis of this neoplasm, discuss pathways that crosstalk with Shh signaling, and the importance of the primary cilium in this neoplastic process. The identification of the basic/translational components of Shh signaling has led to the discovery of potential mechanism-driven druggable targets and subsequent clinical trials have confirmed their remarkable efficacy in treating BCCs, particularly in patients with nevoid BCC syndrome (NBCCS), an autosomal dominant disorder in which patients inherit a germline mutation in the tumor-suppressor gene Patched (Ptch). Patients with NBCCS develop dozens to hundreds of BCCs due to derepression of the downstream G-protein-coupled receptor Smoothened (SMO). Ptch mutations permit transposition of SMO to the primary cilium followed by enhanced expression of transcription factors Glis that drive cell proliferation and tumor growth. Clinical trials with the SMO inhibitor, vismodegib, showed remarkable efficacy in patients with NBCCS, which finally led to its FDA approval in 2012.


Assuntos
Síndrome do Nevo Basocelular/metabolismo , Proteínas Hedgehog/metabolismo , Animais , Síndrome do Nevo Basocelular/tratamento farmacológico , Síndrome do Nevo Basocelular/genética , Síndrome do Nevo Basocelular/patologia , Processos de Crescimento Celular/fisiologia , Modelos Animais de Doenças , Humanos , Receptores de Superfície Celular/metabolismo , Transdução de Sinais
13.
PLoS One ; 9(5): e97245, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24824222

RESUMO

p38 mitogen-activated protein kinases (MAPKs) respond to a wide range of extracellular stimuli. While the inhibition of p38 signaling is implicated in the impaired capacity to repair ultraviolet (UV)-induced DNA damage-a primary risk factor for human skin cancers-its mechanism of action in skin carcinogenesis remains unclear, as both anti-proliferative and survival functions have been previously described. In this study, we utilized cultured keratinocytes, murine tumorigenesis models, and human cutaneous squamous cell carcinoma (SCC) specimens to assess the effect of p38 in this regard. UV irradiation of normal human keratinocytes increased the expression of all four p38 isoforms (α/ß/γ/δ); whereas irradiation of p53-deficient A431 keratinocytes derived from a human SCC selectively decreased p38α, without affecting other isoforms. p38α levels are decreased in the majority of human cutaneous SCCs assessed by tissue microarray, suggesting a tumor-suppressive effect of p38α in SCC pathogenesis. Genetic and pharmacological inhibition of p38α and in A431 cells increased cell proliferation, which was in turn associated with increases in NAPDH oxidase (NOX2) activity as well as intracellular reactive oxygen species (ROS). These changes led to enhanced invasiveness of A431 cells as assessed by the matrigel invasion assay. Chronic treatment of p53-/-/SKH-1 mice with the p38 inhibitor SB203580 accelerated UV-induced SCC carcinogenesis and increased the expression of NOX2. NOX2 knockdown suppressed the augmented growth of A431 xenografts treated with SB203580. These findings indicate that in the absence of p53, p38α deficiency drives SCC growth and progression that is associated with enhanced NOX2 expression and ROS formation.


Assuntos
Carcinoma de Células Escamosas/metabolismo , Regulação Enzimológica da Expressão Gênica/efeitos da radiação , Sistema de Sinalização das MAP Quinases/fisiologia , Espécies Reativas de Oxigênio/metabolismo , Neoplasias Cutâneas/metabolismo , Raios Ultravioleta , Animais , Carcinogênese/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células , Células Cultivadas , Humanos , Imidazóis/efeitos adversos , Queratinócitos/metabolismo , Queratinócitos/efeitos da radiação , Sistema de Sinalização das MAP Quinases/efeitos da radiação , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Knockout , Análise em Microsséries , NADPH Oxidase 2 , NADPH Oxidases/metabolismo , Piridinas/efeitos adversos , Proteína Supressora de Tumor p53/deficiência
14.
J Invest Dermatol ; 134(6): 1579-1588, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24284421

RESUMO

Retinoids are known to affect skin cell proliferation and differentiation and are key molecules that target retinoid and retinoic acid receptors (RXRs and RARs), leading to physiological and pharmacologic effects. Our aim was to elucidate the role of the retinol-binding protein receptor STRA6, mediating cellular uptake of retinol, on skin structure and function. Our results indicate that STRA6 is constitutively expressed in human epidermal keratinocytes and dermal fibroblasts and is regulated via RAR/RXR-mediated pathways. HaCaT (Human adult low Calcium high Temperature) cells with stable STRA6 knockdown (STRA6KD) showed increased proliferation. Consistently, human organotypic 3D skin models using stable STRA6KD HaCaT cells showed a significantly thicker epidermis and enhanced expression of activation, differentiation, and proliferation markers. The effects were reversible after treatment with free retinol. Human skin reconstitution employing STRA6KD HaCaT cells leads to massive epithelial thickening under in vivo conditions in SCID mice. We propose that STRA6KD could lead to cellular vitamin A deficiency in keratinocytes. Consequently, STRA6 has a role for regulating retinoid homeostasis and in helping to program signaling that drives proliferation and differentiation of human skin cells. By its influence on hyperproliferation-associated differentiation, STRA6 could also have a role in skin regeneration and could be a target for pharmacological approaches to improve wound healing.


Assuntos
Epiderme/metabolismo , Regulação da Expressão Gênica , Queratinócitos/metabolismo , Proteínas de Membrana/metabolismo , Animais , Diferenciação Celular , Linhagem Celular , Núcleo Celular/metabolismo , Proliferação de Células , Fibroblastos/metabolismo , Humanos , Queratinócitos/citologia , Ligantes , Camundongos , Camundongos SCID , Receptores do Ácido Retinoico/metabolismo , Regeneração , Pele/metabolismo , Tretinoína/metabolismo , Vitamina A/farmacologia
15.
PLoS One ; 7(6): e39691, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22761871

RESUMO

In order to model squamous cell carcinoma development in vivo, researchers have long preferred hairless mouse models such as SKH-1 mice that have traditionally been classified as 'wild-type' mice irrespective of the genetic factors underlying their hairless phenotype. The work presented here shows that mutations in the Hairless (Hr) gene not only result in the hairless phenotype of the SKH-1 and Hr(-/-) mouse lines but also cause aberrant activation of NFκB and its downstream effectors. We show that in the epidermis, Hr is an early UVB response gene that regulates NFκB activation and thereby controls cellular responses to irradiation. Therefore, when Hr expression is decreased in Hr mutant animals there is a corresponding increase in NFκB activity that is augmented by UVB irradiation. This constitutive activation of NFκB in the Hr mutant epidermis leads to the stimulation a large variety of downstream effectors including the cell cycle regulators cyclin D1 and cyclin E, the anti-apoptosis protein Bcl-2, and the pro-inflammatory protein Cox-2. Therefore, Hr loss results in a state of uncontrolled epidermal proliferation that promotes tumor development, and Hr mutant mice should no longer be considered merely hairless 'wild-type' mice. Instead, Hr is a crucial UVB response gene and its loss creates a permissive environment that potentiates increased tumorigenesis.


Assuntos
Transformação Celular Neoplásica/genética , NF-kappa B/metabolismo , Transdução de Sinais , Fatores de Transcrição/fisiologia , Raios Ultravioleta , Animais , Camundongos , Camundongos Mutantes , Neoplasias Induzidas por Radiação/genética , Fatores de Transcrição/genética , Regulação para Cima
16.
Photochem Photobiol ; 88(5): 1165-72, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22272775

RESUMO

Macroautophagy is a cellular response to various environmental stresses that ensures lysosomal degradation of long-lived and damaged proteins and cellular organelles. It occurs through the formation of an autophagosome, which then fuses with a lysosome to form an autolysosome. Depending on the cellular context, autophagy may promote cancer cell survival or it may serve as a mechanism of tumor suppression. Herein, we show that resveratrol, a natural phytoalexin, induces premature senescence in human A431 SCC cells, and that resveratrol-induced premature senescence is associated with a blockade of autolysosome formation, as assessed by the absence of colocalization of LC3 and Lamp-2, markers for autophagosomes and lysosomes, respectively. Further, we show that resveratrol downregulates the level of Rictor, a component of mTORC2, leading to decreased RhoA-GTPase and altered actin cytoskeleton organization. Exogenous overexpression of Rictor restores RhoA-GTPase activity and actin cytoskeleton network, and decreases resveratrol-induced senescence-associated ß-gal activity, indicating a direct role of Rictor in senescence induction. Rictor is overexpressed in UV-induced murine SCCs, whereas its expression is diminished by oral administration of resveratrol. These data indicate that resveratrol attenuates autophagic process via Rictor, and suggest that downregulation of Rictor may be a mechanism of tumor suppression associated with premature senescence.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Carcinoma de Células Escamosas/tratamento farmacológico , Proteínas de Transporte/antagonistas & inibidores , Neoplasias Cutâneas/tratamento farmacológico , Pele/efeitos dos fármacos , Estilbenos/farmacologia , Animais , Autofagia/efeitos dos fármacos , Autofagia/efeitos da radiação , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patologia , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Linhagem Celular Tumoral , Senescência Celular/efeitos dos fármacos , Senescência Celular/efeitos da radiação , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos da radiação , Humanos , Proteína 2 de Membrana Associada ao Lisossomo/genética , Proteína 2 de Membrana Associada ao Lisossomo/metabolismo , Alvo Mecanístico do Complexo 2 de Rapamicina , Camundongos , Camundongos Pelados , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Complexos Multiproteicos/genética , Complexos Multiproteicos/metabolismo , Fagossomos/efeitos dos fármacos , Fagossomos/efeitos da radiação , Proteína Companheira de mTOR Insensível à Rapamicina , Resveratrol , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/efeitos da radiação , Pele/patologia , Pele/efeitos da radiação , Neoplasias Cutâneas/metabolismo , Neoplasias Cutâneas/patologia , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/metabolismo , Raios Ultravioleta
17.
Cell Cycle ; 10(22): 3849-52, 2011 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-22067472

RESUMO

Deregulated mTOR signaling drives the growth of various human cancers, making mTOR a major target for development of cancer chemotherapeutics. The role of mTOR in carcinogenesis is thought to be largely a consequence of its activity in the cytoplasm resulting in increased translation of pro-tumorigenic genes. However, emerging data locate mTOR in various subcellular compartments including Golgi, mitochondria, endoplasmic reticulum, and the nucleus, implying the presence of compartment-specific mTOR substrates and functions. Efforts to identify mTOR substrates in these compartments, and the mechanisms by which mTOR recruits these substrates and affects downstream cellular processes, will add to our understanding of the diversity of roles played by mTOR in carcinogenesis.


Assuntos
Carcinoma de Células Escamosas/genética , Serina-Treonina Quinases TOR/fisiologia , Motivos de Aminoácidos , Animais , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patologia , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Epigênese Genética , Regulação Neoplásica da Expressão Gênica , Humanos , Camundongos , Transdução de Sinais , Serina-Treonina Quinases TOR/química , Serina-Treonina Quinases TOR/genética
18.
J Biol Chem ; 286(21): 19100-8, 2011 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-21471201

RESUMO

DNA-damaging agents can induce premature senescence in cancer cells, which contributes to the static effects of cancer. However, senescent cancer cells may re-enter the cell cycle and lead to tumor relapse. Understanding the mechanisms that control the viability of senescent cells may be helpful in eliminating these cells before they can regrow. Treating human squamous cell carcinoma (SCC) cells with the anti-cancer compounds, resveratrol and doxorubicin, triggered p53-independent premature senescence by invoking oxidative stress-mediated DNA damage. This process involved the mTOR-dependent phosphorylation of SIRT1 at serine 47, resulting in the inhibition of the deacetylase activity of SIRT1. SIRT1 phosphorylation caused concomitant increases in p65/RelA NF-κB acetylation and the expression of an anti-apoptotic Bfl-1/A1. SIRT1 physically interacts with the mTOR-Raptor complex, and a single amino acid substitution in the TOS (TOR signaling) motif in the SIRT1 prevented Ser-47 phosphorylation and Bfl-1/A1 induction. The pharmacologic and genetic inhibition of mTOR, unphosphorylatable S47A, or F474A TOS mutants restored SIRT1 deacetylase activity, blocked Bfl-1/A1 induction, and sensitized prematurely senescent SCC cells for apoptosis. We further show that the treatment of UVB-induced SCCs with doxorubicin transiently stabilized tumor growth but was followed by tumor regrowth upon drug removal in p53(+/-)/SKH-1 mice. The subsequent treatment of stabilized SCCs with rapamycin decreased tumor size and induced caspase-3 activation. These results demonstrate that the inhibition of SIRT1 by mTOR fosters survival of DNA damage-induced prematurely senescent SCC cells via Bfl-1/A1 in the absence of functional p53.


Assuntos
Carcinoma de Células Escamosas/metabolismo , Senescência Celular , Dano ao DNA , Sirtuína 1/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Acetilação/efeitos dos fármacos , Acetilação/efeitos da radiação , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Substituição de Aminoácidos , Animais , Antibióticos Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Apoptose/genética , Apoptose/efeitos da radiação , Carcinoma de Células Escamosas/genética , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Caspase 3/genética , Caspase 3/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Sobrevivência Celular/efeitos da radiação , Doxorrubicina/farmacologia , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/genética , Ativação Enzimática/efeitos da radiação , Humanos , Camundongos , Camundongos Knockout , Antígenos de Histocompatibilidade Menor , Mutação de Sentido Incorreto , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/genética , Estresse Oxidativo/efeitos da radiação , Fosforilação/efeitos dos fármacos , Fosforilação/genética , Fosforilação/efeitos da radiação , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Proteína Regulatória Associada a mTOR , Sirtuína 1/genética , Serina-Treonina Quinases TOR/genética , Fator de Transcrição RelA/genética , Fator de Transcrição RelA/metabolismo , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Raios Ultravioleta
19.
Exp Dermatol ; 20(5): 450-2, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21366702

RESUMO

Several transport proteins are constitutively expressed in skin cells, but the putative role of the ABC transporter P-glycoprotein (P-gp) in human skin is yet unknown. Therefore, we analysed mRNA and protein expression and localization of P-gp in human skin. Using qRT-PCR, we demonstrated a strong MDR1 mRNA expression in whole skin specimens and dermis, whereas the expression of MDR1 in epidermis, epidermal keratinocytes or dermal fibroblasts was only weak. Immunohistochemistry confirmed mRNA data and revealed a marked expression of P-gp within sweat ducts, vessels, nerve sheaths and muscles of human skin and a moderate expression in basal epidermis. Our findings closely correlate with previous studies in murine skin supporting the role of P-gp in the uptake of compounds from the epidermal compartment and their secretion into the bloodstream and sweat ducts. It may also prevent the uptake of xenobiotics into the skin by functioning as a barrier located in the dermal vasculature.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/genética , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Derme/metabolismo , Expressão Gênica/genética , Pele/metabolismo , Subfamília B de Transportador de Cassetes de Ligação de ATP , Vasos Sanguíneos/metabolismo , Carcinoma de Células Escamosas/metabolismo , Células Cultivadas , Epiderme/metabolismo , Fibroblastos/metabolismo , Humanos , Queratinócitos/metabolismo , Fígado/metabolismo , Músculo Liso/metabolismo , Bainha de Mielina/metabolismo , Pele/irrigação sanguínea , Neoplasias Cutâneas/metabolismo , Glândulas Sudoríparas/metabolismo
20.
J Invest Dermatol ; 131(1): 195-202, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20720562

RESUMO

Resveratrol (RES) is a potent anti-cancer agent. We have previously reported that RES arrests the growth of invasive human A431 squamous cell carcinoma (SCC) cells. In this study, we show that oral administration of RES to highly tumor-susceptible p53(+/-)/SKH-1 mice markedly delayed UV-induced skin tumorigenesis and reduced the malignant conversion of benign papillomas to SCCs. Transforming growth factor-ß2 (TGF-ß2) was predominantly overexpressed in UV-induced SCCs and its expression was diminished in RES-treated SCCs/skin. In addition to the inhibition of TGF-ß2 expression, RES increased the level of epithelial cadherin. This RES-mediated TGF-ß2 downregulation led to the inhibition of both TGF-ß2/Smad-dependent and -independent pathways, and suppressed the invasiveness of A431 cells. Addition of TGF-ß2, but not TGF-ß1, rescued the RES-mediated downregulation of p-extracellular signal-regulated kinases 1/2, p-Smad3, and α-smooth muscle actin. The protein kinase B (Akt) substrate cAMP response-binding protein (pCREB) transcription factor is known to regulate TGF-ß2 expression, and RES treatment decreased phosphorylation of Akt and pCREB. Expression of constitutively active Akt blocked RES inhibition of CREB and TGF-ß2, and rescued RES inhibition of cellular invasiveness. Our data indicate that RES suppresses UV-induced malignant tumor progression in p53(+/-)/SKH-1 mice and that RES-inhibited invasiveness of human A431 SCC cells appears to occur, in part, through the Akt-mediated downregulation of TGF-ß2.


Assuntos
Anticarcinógenos/farmacologia , Carcinoma de Células Escamosas , Neoplasias Cutâneas , Estilbenos/farmacologia , Fator de Crescimento Transformador beta2/metabolismo , Raios Ultravioleta/efeitos adversos , Animais , Carcinoma de Células Escamosas/tratamento farmacológico , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patologia , Linhagem Celular Tumoral , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/fisiologia , Regulação para Baixo/efeitos da radiação , Feminino , Humanos , Masculino , Camundongos , Camundongos Pelados , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Resveratrol , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Transdução de Sinais/efeitos da radiação , Neoplasias Cutâneas/tratamento farmacológico , Neoplasias Cutâneas/metabolismo , Neoplasias Cutâneas/patologia , Proteína Supressora de Tumor p53/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...