Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell Biol Int ; 46(5): 737-746, 2022 May.
Artigo em Inglês | MEDLINE | ID: mdl-35032143

RESUMO

Hair follicles regenerate periodically by spontaneously undergoing cycles of growth, regression, and relative quiescence. During the hair cycle, follicle stem cells residing in a specialized niche remain quiescent, and they are stimulated to proliferate throughout the growth phase of the hair follicle. Although cell cycle regulators play a prominent role during the activation of hair follicle stem cells, the identity and the role of these regulators have not been confirmed. Herein, we reported that stem cells located in the bulge region of the HF (BuSCs) express high levels of cyclin-dependent kinase 4 (CDK4) through the quiescent phase of the hair cycle. Using gain- and loss-of-function studies, we have determined that the CDK4 protein level affects the number of BuSCs. Transgenic expression of CDK4 in the bulge region of the hair follicles reduces the number of BuSCs, whereas CDK4 ablation resulted in an increasing number of BuSCs. These results suggest that deregulation of CDK4 protein levels contributes to distorting the self-renewal/proliferation balance and, in turn, altering the number of BuSCs.


Assuntos
Quinase 4 Dependente de Ciclina/metabolismo , Folículo Piloso , Células-Tronco , Animais , Folículo Piloso/metabolismo , Queratinócitos , Camundongos
2.
Oncol Lett ; 22(4): 732, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34429772

RESUMO

Cyclin-dependent kinase 4 (CDK4) is a critical molecule that regulates key aspects of cell proliferation through phosphorylation of the retinoblastoma (Rb) family of proteins. In the last few years, it has been suggested that CDK4 plays alternative roles in cell proliferation and tumorigenesis. The main aim of the present study was to define a novel CDK4 function as a transcriptional regulator of genes involved in chromosome segregation, contributing to the G2/M phase transition. Herein, chromatin-immunoprecipitation reverse transcription-quantitative PCR assays were performed to demonstrate that CDK4 could occupy the promoter region of genes associated with chromosomal segregation, such as Aurora-B (Aurkb) and Centromere Protein P (CENP-P). Moreover, gain- and loss-of-function experiments showed that CDK4 participated in the transcriptional regulation of Aurkb and CENP-P. The finding that Aurkb may have a crucial role in chromosome bi-orientation and the spindle assembly checkpoint, and that CENP-P could be required for proper kinetochore function suggests that dysregulation of CDK4 expression induces chromosomal instability and, in some cases, cancer development.

3.
Oncol Lett ; 14(6): 6497-6504, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29181098

RESUMO

A subset of cells, termed side-population (SP), which have the ability to efflux Hoeschst 33342, have previously been demonstrated to act as a potential method to isolate stem cells. Numerous stem/progenitor cells have been localized in different regions of the mouse hair follicle (HF). The present study identified a SP in the mouse HF expressing the ABCG2 transporter and MTS24 surface marker. These cells are restricted to the upper isthmus of the HF and have previously been described as progenitor cells. Consistent with their SP characteristic, they demonstrated elevated expression of ABCG2 transporter, which participates in the dye efflux. Analysis of tumor epidermal cell lines revealed a correlation between the number of SP keratinocytes and the grade of malignancy, suggesting that the SP may play a role in malignant progression. Consistent with this idea, the present study observed an increased number of cells expressing ABCG2 and MTS24 in chemically induced skin tumors and skin tumor cell lines. This SP does not express the CD34 surface marker detected in the multipotent stem cells of the bulge region of the HF, which have been defined as tumor initiation cells. The present study concluded that a SP with properties of progenitor cells is localized in the upper isthmus of the HF and is important in mouse skin tumor progression.

4.
Oncol Lett ; 14(3): 2723-2734, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28927034

RESUMO

Rearrangement and amplification of the D-type cyclin genes have been reported in human cancer. Previous studies have demonstrated that Ras-mediated skin tumorigenesis depends on pathways that act through cyclin D1 and D2; however, the role of cyclin D3 remains unknown. The present study demonstrates that cyclin D3 ablation does not affect keratinocyte proliferation, but instead increases apoptosis levels in the bulge region of the hair follicle. Consequently, cyclin D3 ablation reduces skin papilloma development in a Ras-dependent carcinogenesis model. Previous results revealed that cyclin D3 preferentially binds to cyclin-dependent kinase 6 (CDK6) in mouse keratinocytes and transgenic expression of CDK6 (K5CDK6 mice) inhibits skin tumor development. Thus, we hypothesized that the inhibitory effect of CDK6 is dependent on cyclin D3 expression. To test this hypothesis, a mouse model that overexpresses CDK6 and does not express cyclin D3 (K5CDK6/cyclin D3-/- compound mouse) was developed. Biochemical analysis of the epidermis of K5CDK6/cyclin D3-/- mice revealed that cyclin D3 ablation resulted in increased expression of cyclin D1 protein, with a consequent elevation in the level of CDK6/cyclin D1 and CDK4/cyclin D1 complexes. These findings were concurrent with the increase skin papilloma malignant progression observed in K5CDK6/cyclin D3-/- mice. In summary the absence of cyclin D3 led to fewer number of papillomas in cyclin D3-ablated mice than in the wild-type owing to increased apoptosis, suggesting that alterations in cell survival are a crucial mechanism for crippling cellular defense against neoplasia. The results of the current study also suggest that although cyclin D3 expression does not alter the tumor suppressive role of CDK6 in skin carcinogenesis, the compensatory increase in cyclin D1 can shift the balance towards malignant progression.

5.
Methods Mol Biol ; 1195: 259-68, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24497321

RESUMO

The mouse skin is composed of at least three differentiating epithelial compartments: the epidermis, the hair follicle, and the associated glands such as the sebaceous glands. Proliferation of these epithelial cells takes place in the keratinocytes' layer or basal cell layer; in the periphery of the sebaceous gland (the basal layer of the gland) and in specific cell compartments around the hair follicle. In mouse skin, an epithelial stem cell population is thought to localize to the bulge region of the hair follicle, a segment that does not undergo regression during the hair cycle. In addition, several other putative stem cells and/or progenitors have been identified in different regions of the hair follicle. Using the Hoeschst exclusion technique, originally described in the hematopoietic system, it has been possible to isolate a mouse keratinocyte cell population with characteristics of stem cells (side-population, SP). One of the main features of these SP is their ability to efflux antimitotic drugs as well as some specific dyes. This characteristic allows for SP cells to be isolated based upon their capacity to efflux the dye Hoechst 33342, through a mechanism driven by a membrane transporter, the breast cancer resistance protein (BCRP1/ABCG2). In this chapter, we described the isolation of SP stem cells from adult mouse hair follicles utilizing the Hoeschst exclusion technique by flow cytometry analysis.


Assuntos
Citometria de Fluxo/métodos , Folículo Piloso/citologia , Células da Side Population/citologia , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/genética , Animais , Regulação da Expressão Gênica , Camundongos , Células da Side Population/metabolismo
6.
Am J Pathol ; 182(5): 1854-64, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23474082

RESUMO

S-phase kinase-associated protein 2 (Skp2) functions as the receptor component of the Skp-Cullin-F-box complex and is implicated in the degradation of several cell cycle regulators, such as p21(Cip1), p27(Kip1), p57(Kip2), and cyclin E. Numerous studies in human and experimental tumors have demonstrated low p27(Kip1) levels and elevated Skp2 expression. However, a direct association between the inverse correlation of Skp2 and p27(Kip1) with tumorigenesis has not been demonstrated. Herein, we provide evidence that skin tumorigenesis is inhibited in Skp2(-/-) mice. An analysis of mouse keratinocytes indicates that increased p27(Kip1) levels in Skp2(-/-) epidermis cause reduced cell proliferation that is alleviated in the epidermis from Skp2(-/-)/p27(-/-) compound mice. In contrast, we establish that a p27(Kip1) deficiency does not overturn the reduced skin tumorigenesis experienced by Skp2(-/-) mice. In addition, Skp2(-/-) epidermis exhibits an accumulation of p53-cofactor CBP/p300 that is associated with elevated apoptosis in hair follicles and decreased skin tumorigenesis. We conclude that p27(Kip1) accumulation is responsible for the hypoplasia observed in normal tissues of Skp2(-/-) mice but does not have a preponderant function in reducing skin tumorigenesis.


Assuntos
Carcinogênese/patologia , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Proteínas Quinases Associadas a Fase S/metabolismo , Neoplasias Cutâneas/patologia , Animais , Apoptose , Carcinogênese/metabolismo , Ciclo Celular , Epiderme/metabolismo , Epiderme/patologia , Deleção de Genes , Humanos , Cinética , Camundongos , Camundongos Endogâmicos C57BL , Papiloma/metabolismo , Papiloma/patologia , Neoplasias Cutâneas/metabolismo , Proteína Supressora de Tumor p53/deficiência , Proteína Supressora de Tumor p53/metabolismo
7.
Cell Cycle ; 11(2): 335-42, 2012 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-22214766

RESUMO

We have previously demonstrated that ras-mediated skin tumorigenesis depends on signaling pathways that act preferentially through cyclin D1 and D2. Interestingly, the expression of cyclin D3 inhibits skin tumor development, an observation that conflicts with the oncogenic role of D-type cyclins in the mouse epidermis. Here, we show that simultaneous up and downregulation of particular members of the D-type cyclin family is a valuable approach to reduce skin tumorigenesis. We developed the K5D3/cyclin D1(-/-) compound mouse, which overexpresses cyclin D3 but lacks expression of cyclin D1 in the skin. Similar to K5D3 transgenic mice, keratinocytes from K5D3/cyclin D1(-/-) compound mice show a significant reduction of cyclin D2 levels. Therefore, this model allows us to determine the effect of cyclin D3 expression when combined with reduced or absent expression of the remaining two members of the D-type cyclin family in mouse epidermis. Our data show that induced expression of cyclin D3 compensates for the reduced level of cyclin D1 and D2, resulting in normal keratinocyte proliferation. However, simultaneous ablation of cyclin D1 and downregulation of cyclin D2 via cyclin D3 expression resulted in a robust reduction in ras-mediated skin tumorigenesis. We conclude that modulation of the levels of particular members of the D-type cyclin family could be useful to inhibit tumor development and, in particular, ras-mediated tumorigenesis.


Assuntos
Carcinoma de Células Escamosas/metabolismo , Ciclina D1/metabolismo , Ciclina D3/metabolismo , Papiloma/metabolismo , Neoplasias Cutâneas/metabolismo , 9,10-Dimetil-1,2-benzantraceno , Animais , Carcinoma de Células Escamosas/induzido quimicamente , Carcinoma de Células Escamosas/patologia , Proliferação de Células , Transformação Celular Neoplásica , Ciclina D1/genética , Ciclina D2/metabolismo , Ciclina D3/genética , Regulação da Expressão Gênica , Camundongos , Camundongos Transgênicos , Proteína Oncogênica p21(ras)/genética , Papiloma/induzido quimicamente , Papiloma/patologia , Pele/patologia , Neoplasias Cutâneas/induzido quimicamente , Neoplasias Cutâneas/patologia , Carga Tumoral
8.
Am J Pathol ; 178(6): 2470-7, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21641375

RESUMO

The proto-oncogene c-Myc encodes a transcription factor that is implicated in the regulation of cellular proliferation, differentiation, and apoptosis. Myc accelerates the rate of cell proliferation, at least in part, through its ability to down-regulate the expression of the cell cycle inhibitor p27(Kip1). Moreover, p27(Kip1) protein levels are regulated by ubiquitin-mediated turnover, leading to destruction by the E3 ubiquitin ligase SCF(Skp2). Therefore, we hypothesize that a lack of Skp2 expression should lead to increased p27(Kip1) levels and further inhibition of Myc-mediated proliferation and tumorigenesis. Myc expression in epithelial tissues of transgenic mice (K5-Myc) led to increased keratinocyte proliferation and the development of spontaneous tumors within the oral cavity. We generated K5-Myc-transgenic mice in an Skp2-null background. Consistent with our hypothesis, we found that Myc-mediated keratinocyte hyperproliferation was abolished by the loss of Skp2. However, Skp2 ablation did not affect Myc-driven tumorigenesis because the incidence, latency, and degree of differentiation of oral tumors were identical between K5-Myc/Skp2(+/+) and K5-Myc/Skp2(-/-) mice. Altogether, these findings suggest that Skp2 and p27(Kip1) are critical for Myc-driven keratinocyte proliferation; however, Myc-mediated tumorigenesis in the oral epithelium is independent of the Skp2-p27(Kip1) axis.


Assuntos
Epitélio/metabolismo , Epitélio/patologia , Queratinócitos/metabolismo , Queratinócitos/patologia , Boca/patologia , Proteínas Proto-Oncogênicas c-myc/metabolismo , Proteínas Quinases Associadas a Fase S/metabolismo , Animais , Proteínas de Ciclo Celular/metabolismo , Proliferação de Células , Epiderme/metabolismo , Epiderme/patologia , Camundongos , Camundongos Transgênicos , Neoplasias Bucais/metabolismo , Neoplasias Bucais/patologia , Fenótipo , Lesões Pré-Cancerosas/patologia
9.
Am J Pathol ; 178(1): 345-54, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21224071

RESUMO

Cyclin-dependent kinases (CDKs) 4 and 6 are important regulators of the G(1) phase of the cell cycle, share 71% amino acid identity, and are expressed ubiquitously. As a result, it was assumed that each of these kinases plays a redundant role regulating normal and neoplastic proliferation. In previous reports, we have described the effects of CDK4 expression in transgenic mice, including the development of epidermal hyperplasia and increased malignant progression to squamous cell carcinoma. To study the role of CDK6 in epithelial growth and tumorigenesis, we generated transgenic mice carrying the CDK6 gene under the keratin 5 promoter (K5CDK6). Similar to K5CDK4 mice, epidermal proliferation increased substantially in K5CDK6 mice; however, no hyperplasia was observed. CDK6 overexpression also triggered keratinocyte apoptosis in interfollicular and follicular epidermis as a compensatory mechanism to override aberrant proliferation. Unexpectedly, CDK6 overexpression results in decreased skin tumor development compared with wild-type siblings. The inhibition in skin tumorigenesis was similar to that previously reported in K5-cyclin D3 mice. Furthermore, biochemical analysis of the K5CDK6 epidermis showed preferential complex formation between CDK6 and cyclin D3, suggesting that this particular complex plays an important role in tumor restraint. These studies provide in vivo evidence that CDK4 and CDK6 play a similar role as a mediator of keratinocyte proliferation but differ in apoptosis activation and skin tumor development.


Assuntos
Transformação Celular Neoplásica/patologia , Quinase 6 Dependente de Ciclina/biossíntese , Epiderme/patologia , Queratinócitos/patologia , Neoplasias Cutâneas/metabolismo , Animais , Proliferação de Células , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Quinase 4 Dependente de Ciclina/biossíntese , Quinase 4 Dependente de Ciclina/genética , Quinase 6 Dependente de Ciclina/genética , Epiderme/enzimologia , Hiperplasia/enzimologia , Queratina-5/genética , Queratinócitos/enzimologia , Camundongos , Camundongos Transgênicos , Regiões Promotoras Genéticas , Neoplasias Cutâneas/enzimologia , Neoplasias Cutâneas/genética
10.
Fitoterapia ; 82(3): 414-21, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21129455

RESUMO

In the present study, we evaluated the effects of extracts and purified compounds from fresh leaves of Rosmarinus officinalis L. Pretreatment with the major anti-inflammatory compounds, carnosic acid (CA) and carnosol (CS), inhibited phorbol 12-myristate 13-acetate (PMA)-induced ear inflammation in mice with an EC(50) of 10.20 µg/cm(2) and 10.70 µg/cm(2), respectively. To further understand the anti-inflammatory mechanism of these compounds, we analyzed the in vivo expression of several inflammation-associated genes in mouse skin by reverse transcriptase-polymerase chain reaction (RT-PCR). Our data showed that CA and CS reduced the expression of IL-1ß and TNF-α but had less effect on fibronectin and ICAM-1 expression. Interestingly, both compounds selectively inhibited COX-2 but not COX-1. Histopathological analysis of hematoxylin and eosin (H&E)-stained tissue revealed a marked reduction in leukocyte infiltration and epidermal ulceration of PMA-treated ears when ears were pretreated with ethanolic extracts or pure CA. In vitro, we showed that ethanolic extract, carnosic acid and carnosol significantly inhibited the overproduction of nitric oxide (NO) in a dose-dependent manner in the RAW 264.7 murine macrophage cell line. For the first time in vivo, we showed that CA and CS differentially regulate the expression of inflammation-associated genes, thus demonstrating the pharmacological basis for the anti-inflammatory properties reported for CA and CS.


Assuntos
Abietanos/uso terapêutico , Inflamação/tratamento farmacológico , Fitoterapia , Extratos Vegetais/uso terapêutico , Rosmarinus/química , Pele/efeitos dos fármacos , Abietanos/farmacologia , Animais , Inibidores de Ciclo-Oxigenase 2/farmacologia , Inibidores de Ciclo-Oxigenase 2/uso terapêutico , Relação Dose-Resposta a Droga , Fibronectinas/genética , Fibronectinas/metabolismo , Expressão Gênica , Inflamação/induzido quimicamente , Inflamação/metabolismo , Molécula 1 de Adesão Intercelular/genética , Molécula 1 de Adesão Intercelular/metabolismo , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Infiltração de Neutrófilos/efeitos dos fármacos , Óxido Nítrico/biossíntese , Extratos Vegetais/farmacologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Pele/metabolismo , Pele/patologia , Acetato de Tetradecanoilforbol , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo
11.
Mol Carcinog ; 48(6): 508-16, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18942117

RESUMO

Cyclin D1b is an alternative transcript of the cyclin D1 gene (CCND1) expressed in human tumors. Its abundance is regulated by a single base pair polymorphism at the exon 4/intron 4 boundary (nucleotide 870). Epidemiological studies have shown a correlation between the presence of the G870A allele (that favors the splicing for cyclin D1b) with increased risk and less favorable outcome in several forms of cancer. More recently, it has been shown that, unlike cyclin D1a, the alternative transcript D1b by itself has the capacity to transform fibroblasts in vitro. In order to study the oncogenic potential of cyclin D1b, we developed transgenic mice expressing human cyclin D1b under the control of the bovine K5 promoter (K5D1b mice). Seven founders were obtained and none of them presented any significant phenotype or developed spontaneous tumors. Interestingly, K5D1b mice do not develop the fatal thymic hyperplasia, which is characteristic of the cyclin D1a transgenic mice (K5D1a). Susceptibility to skin carcinogenesis was tested in K5D1b mice using two-stage carcinogenesis protocols. In two independent experiments, K5D1b mice developed higher papilloma multiplicity as compared with wild-type littermates. However, when K5D1b mice were crossed with cyclin D1KO mice, the expression of cyclin D1b was unable to rescue the carcinogenesis-resistant phenotype of the cyclin D1 KO mice. To further explore the role of cyclin D1b in mouse models of carcinogenesis we carried out in silico analysis and in vitro experiments to evaluate the existence of a mouse homologous of the human cyclin D1b transcript. We were unable to find any evidence of an alternatively spliced transcript in mouse Ccnd1. These results show that human cyclin D1b has different biological functions than cyclin D1a and confirm its oncogenic properties.


Assuntos
Transformação Celular Neoplásica/genética , Ciclina D1/genética , Neoplasias Cutâneas/genética , Timo/patologia , Animais , Sequência de Bases , Primers do DNA , Éxons , Hiperplasia , Íntrons , Camundongos , Camundongos Transgênicos , Reação em Cadeia da Polimerase , Polimorfismo de Nucleotídeo Único
12.
Am J Pathol ; 173(2): 526-35, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18599613

RESUMO

It has been widely assumed that elevated CDK2 kinase activity plays a contributory role in tumorigenesis. We have previously shown that mice overexpressing CDK4 under control of the keratin 5 promoter (K5CDK4 mice) develop epidermal hyperplasia and increased susceptibility to squamous cell carcinomas. In this model, CDK4 overexpression results in increased CDK2 activity associated with the noncatalytic function of CDK4, sequestration of p21(Cip1) and p27(Kip1). Furthermore, we have shown that ablation of Cdk2 reduces Ras-Cdk4 tumorigenesis, suggesting that increased CDK2 activity plays an important role in Ras-mediated tumorigenesis. To investigate this hypothesis, we generated two transgenic mouse models of elevated CDK2 kinase activity, K5Cdk2 and K5Cdk4(D158N) mice. The D158N mutation blocks CDK4 kinase activity without interfering with its binding capability. CDK2 activation via overexpression of CDK4(D158N), but not of CDK2, resulted in epidermal hyperplasia. We observed elevated levels of p21(Cip1) in K5Cdk2, but not in K5Cdk4(D158N), epidermis, suggesting that CDK2 overexpression elicits a p21(Cip1) response to maintain keratinocyte homeostasis. Surprisingly, we found that neither CDK2 overexpression nor the indirect activation of CDK2 enhanced skin tumor development. Thus, although the indirect activation of CDK2 is sufficient to induce keratinocyte hyperproliferation, activation of CDK2 alone does not induce malignant progression in Ras-mediated tumorigenesis.


Assuntos
Proliferação de Células , Transformação Celular Neoplásica/metabolismo , Quinase 2 Dependente de Ciclina/metabolismo , Epiderme/enzimologia , Queratinócitos/patologia , Neoplasias Cutâneas/enzimologia , 9,10-Dimetil-1,2-benzantraceno , Animais , Transformação Celular Neoplásica/patologia , Quinase 2 Dependente de Ciclina/genética , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Ativação Enzimática , Epiderme/patologia , Queratinócitos/enzimologia , Camundongos , Camundongos Transgênicos , Mutação , Neoplasias Cutâneas/induzido quimicamente , Neoplasias Cutâneas/patologia
13.
Cancer Res ; 68(1): 162-71, 2008 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-18172308

RESUMO

The keratin 5 (K5) promoter drives transgenic expression to the basal cell layer of stratified epithelia. Surprisingly, analysis of K5CDK4 and K5CDK2 transgenic mouse embryos showed CDK4 and CDK2 expression not only in the expected tissues, but also in the adenohypophysis. This organ is derived from an upwards growth of the primitive oropharynx, a K5-expressing tissue. We show that transgenic expression of CDKs in the embryonic oral ectoderm is specifically retained in undifferentiated cells from the pars intermedia of the adenohypophysis. Interestingly, we found that K5CDK4 mice show a decreased number of pituitary stem cells, even though CDK4 is not expressed in the stem cells but in transit-amplifying (TA)-like cells. Interestingly, CDK4-expressing cells, but not CDK2-expressing cells, strongly synergize with lack of p27(Kip1) to generate pituitary carcinomas that appear with shortened latency and are drastically more aggressive than those arising in p27(-/-) mice. Thus, we show that deregulation of CDK expression in the primitive oral epithelium plays a unique function, providing a selective advantage that gives rise to transgene-positive TA-like pituitary cells. Furthermore, retention of CDK4 in these TA-like pituitary cells synergizes with loss of p27(Kip1) to induce pituitary adenocarcinomas. This model suggests that forced expression of CDK4 sensitizes cells and synergizes with a second change resulting in tumor development.


Assuntos
Transformação Celular Neoplásica/metabolismo , Quinase 2 Dependente de Ciclina/metabolismo , Quinase 4 Dependente de Ciclina/metabolismo , Boca/enzimologia , Hipófise/enzimologia , Neoplasias Hipofisárias/enzimologia , Animais , Contagem de Células , Transformação Celular Neoplásica/genética , Quinase 2 Dependente de Ciclina/análise , Quinase 2 Dependente de Ciclina/genética , Quinase 4 Dependente de Ciclina/análise , Quinase 4 Dependente de Ciclina/genética , Inibidor de Quinase Dependente de Ciclina p27/genética , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Humanos , Camundongos , Camundongos Transgênicos , Boca/embriologia , Neoplasias Hipofisárias/genética , Regiões Promotoras Genéticas , Células-Tronco
14.
Cancer Res ; 67(20): 9713-20, 2007 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-17942901

RESUMO

We have previously shown that forced expression of CDK4 in mouse skin (K5CDK4 mice) results in increased susceptibility to squamous cell carcinoma (SCC) development in a chemical carcinogenesis protocol. This protocol induces skin papilloma development, causing a selection of cells bearing activating Ha-ras mutations. We have also shown that myc-induced epidermal proliferation and oral tumorigenesis (K5Myc mice) depends on CDK4 expression. Biochemical analysis of K5CDK4 and K5Myc epidermis as well as skin tumors showed that keratinocyte proliferation is mediated by CDK4 sequestration of p27Kip1 and p21Cip1, and activation of CDK2. Here, we studied the role of CDK2 in epithelial tumorigenesis. In normal skin, loss of CDK2 rescues CDK4-induced, but not myc-induced epidermal hyperproliferation. Ablation of CDK2 in K5CDK4 mice results in decreased incidences and multiplicity of skin tumors as well as malignant progression to SCC. Histopathologic analysis showed that K5CDK4 tumors are drastically more aggressive than K5CDK4/CDK2-/- tumors. On the other hand, we show that CDK2 is dispensable for myc-induced tumorigenesis. In contrast to our previous report of K5Myc/CDK4-/-, K5Myc/CDK2-/- mice developed oral tumors with the same frequency as K5Myc mice. Overall, we have established that ras-induced tumors are more susceptible to CDK2 ablation than myc-induced tumors, suggesting that the efficacy of targeting CDK2 in tumor development and malignant progression is dependent on the oncogenic pathway involved.


Assuntos
Carcinoma de Células Escamosas/patologia , Transformação Celular Neoplásica/patologia , Quinase 2 Dependente de Ciclina/deficiência , Quinase 4 Dependente de Ciclina/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Neoplasias Cutâneas/patologia , Proteínas ras/metabolismo , Animais , Carcinógenos , Carcinoma de Células Escamosas/enzimologia , Carcinoma de Células Escamosas/metabolismo , Transformação Celular Neoplásica/metabolismo , Quinase 2 Dependente de Ciclina/metabolismo , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Progressão da Doença , Ativação Enzimática , Feminino , Masculino , Camundongos , Camundongos Transgênicos , Proteínas Proto-Oncogênicas c-myc/biossíntese , Proteínas Proto-Oncogênicas c-myc/genética , Neoplasias Cutâneas/induzido quimicamente , Neoplasias Cutâneas/enzimologia , Acetato de Tetradecanoilforbol
15.
Mol Cell Biol ; 24(17): 7538-47, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15314163

RESUMO

The proto-oncogene c-myc encodes a transcription factor that is implicated in the regulation of cellular proliferation, differentiation, and apoptosis and that has also been found to be deregulated in several forms of human and experimental tumors. We have shown that forced expression of c-myc in epithelial tissues of transgenic mice (K5-Myc) resulted in keratinocyte hyperproliferation and the development of spontaneous tumors in the skin and oral cavity. Although a number of genes involved in cancer development are regulated by c-myc, the actual mechanisms leading to Myc-induced neoplasia are not known. Among the genes regulated by Myc is the cyclin-dependent kinase 4 (CDK4) gene. Interestingly, previous studies from our laboratory showed that the overexpression of CDK4 led to keratinocyte hyperproliferation, although no spontaneous tumor development was observed. Thus, we tested the hypothesis that CDK4 may be one of the critical downstream genes involved in Myc carcinogenesis. Our results showed that CDK4 inhibition in K5-Myc transgenic mice resulted in the complete inhibition of tumor development, suggesting that CDK4 is a critical mediator of tumor formation induced by deregulated Myc. Furthermore, a lack of CDK4 expression resulted in marked decreases in epidermal thickness and keratinocyte proliferation compared to the results obtained for K5-Myc littermates. Biochemical analysis of the K5-Myc epidermis showed that CDK4 mediates the proliferative activities of Myc by sequestering p21Cip1 and p27Kip1 and thereby indirectly activating CDK2 kinase activity. These results show that CDK4 mediates the proliferative and oncogenic activities of Myc in vivo through a mechanism that involves the sequestration of specific CDK inhibitors.


Assuntos
Quinases Ciclina-Dependentes/metabolismo , Epitélio/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Proteínas Proto-Oncogênicas , Neoplasias Cutâneas/metabolismo , Animais , Ciclo Celular/fisiologia , Proteínas de Ciclo Celular/metabolismo , Divisão Celular/fisiologia , Quinase 4 Dependente de Ciclina , Inibidor de Quinase Dependente de Ciclina p21 , Inibidor de Quinase Dependente de Ciclina p27 , Quinases Ciclina-Dependentes/antagonistas & inibidores , Quinases Ciclina-Dependentes/genética , Ciclinas/metabolismo , Células Epidérmicas , Epiderme/metabolismo , Epiderme/patologia , Epitélio/patologia , Humanos , Queratinócitos/citologia , Queratinócitos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fenótipo , Proto-Oncogene Mas , Proteínas Proto-Oncogênicas c-myc/genética , Neoplasias Cutâneas/genética , Proteínas Supressoras de Tumor/metabolismo
16.
Oncogene ; 23(10): 1863-73, 2004 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-14647432

RESUMO

In a previous study, we reported that overexpression of cyclin-dependent kinase-4 (CDK4) in mouse epidermis results in epidermal hyperplasia, hypertrophy and severe dermal fibrosis. In this study, we have investigated the susceptibility to skin tumor formation by forced expression of CDK4. Skin tumors from transgenic mice showed a dramatic increase in the rate of malignant progression to squamous cell carcinomas (SCC) in an initiation-promotion protocol. Histopathological analysis of papillomas from transgenic mice showed an elevated number of premalignant lesions characterized by dysplasia and marked atypia. Interestingly, transgenic mice also developed tumors in initiated but not promoted skin, demonstrating that CDK4 replaced the action of tumor promoters. These results suggest that expression of cyclin D1 upon ras activation synergizes with CDK4 overexpression. However, cyclin D1 transgenic mice and double transgenic mice for cyclin D1 and CDK4 did not show increased malignant progression in comparison to CDK4 transgenic mice. Biochemical analysis of tumors showed that CDK4 sequesters the CDK2 inhibitors p27Kip1 and p21Cip1, suggesting that indirect activation of CDK2 plays an important role in tumor development. These results indicate that, contrary to the general assumption, the catalytic subunit, CDK4, has higher oncogenic activity than cyclin D1, revealing a potential use of CDK4 as therapeutic target.


Assuntos
Quinases Ciclina-Dependentes/genética , Proteínas Proto-Oncogênicas , Neoplasias Cutâneas/genética , Animais , Carcinoma/genética , Carcinoma/patologia , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patologia , Quinase 4 Dependente de Ciclina , Humanos , Camundongos , Camundongos Transgênicos , Papiloma/genética , Papiloma/patologia , Neoplasias Cutâneas/patologia
17.
Am J Pathol ; 161(2): 405-11, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12163365

RESUMO

Most human tumors have mutations that result in deregulation of the cdk4/cyclin-Ink4-Rb pathway. Overexpression of D-type cyclins or cdk4 and inactivation of Ink4 inhibitors are common in human tumors. Conversely, lack of cyclin D1 expression results in significant reduction in mouse skin and mammary tumor development. However, complete elimination of tumor development was not observed in these models, suggesting that other cyclin/cdk complexes play an important role in tumorigenesis. Here we described the effects of cdk4 deficiency on mouse skin proliferation and tumor development. Cdk4 deficiency resulted in a 98% reduction in the number of tumors generated through the two-stage carcinogenesis model. The absence of cdk4 did not affect normal keratinocyte proliferation and both wild-type and cdk4 knockout epidermis are equally affected after topical treatment with the tumor promoter 12-O-tetradecanoylphorbol-13-acetate (TPA), resulting in epidermal hyperplasia. In similar fashion, cdk4 knockout keratinocytes proliferated well in an in vivo model of wound-induced proliferation. Biochemical studies in mouse epidermis showed that cdk6 activity increased twofold in cdk4-deficient mice compared to wild-type siblings. These results suggest that therapeutic approaches to inhibit cdk4 activity could provide a target to inhibit tumor development with minimal or no effect in normal tissue.


Assuntos
Quinases Ciclina-Dependentes/genética , Queratinócitos/citologia , Proteínas Proto-Oncogênicas , Neoplasias Cutâneas/genética , Animais , Carcinógenos/toxicidade , Divisão Celular/genética , Transformação Celular Neoplásica/genética , Quinase 4 Dependente de Ciclina , Quinases Ciclina-Dependentes/deficiência , Queratinócitos/enzimologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neoplasias Cutâneas/enzimologia , Neoplasias Cutâneas/etiologia , Acetato de Tetradecanoilforbol/toxicidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...