Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
J Invest Dermatol ; 142(11): 2873-2884.e7, 2022 11.
Article in English | MEDLINE | ID: mdl-35551922

ABSTRACT

Altered fibroblast GF receptor (FGFR) signaling has been shown to play a role in a number of cancers. However, the role of FGFR signaling in the development and progression of UVB-induced cutaneous squamous cell carcinoma remains unclear. In this study, the effect of UVB radiation on FGFR activation and its downstream signaling in mouse skin epidermis was examined. In addition, the impact of FGFR inhibition on UVB-induced signaling and skin carcinogenesis was also investigated. Exposure of mouse dorsal skin to UVB significantly increased the phosphorylation of FGFRs in the epidermis as well as the activation of downstream signaling pathways, including protein kinase B/mTOR, signal transducers and activators of transcription, and MAPK. Topical application of the pan-FGFR inhibitor AZD4547 to mouse skin before exposure to UVB significantly inhibited FGFR phosphorylation as well as mTORC1, signal transducer and activator of transcription 3, and MAPK activation (i.e., phosphorylation). Moreover, AZD4547 pretreatment significantly inhibited UVB-induced epidermal hyperplasia and hyperproliferation and reduced the infiltration of mast cells and macrophages into the dermis. AZD4547 treatment also significantly inhibited mRNA expression of inflammatory genes in the epidermis. Finally, mice treated topically with AZD4547 before UVB exposure showed decreased cutaneous squamous cell carcinoma incidence and increased survival rate. Collectively, the current data support the hypothesis that inhibition of FGFR in the epidermis may provide a new strategy to prevent and/or treat UVB-induced cutaneous squamous cell carcinoma.


Subject(s)
Carcinoma, Squamous Cell , Skin Neoplasms , Mice , Animals , Receptors, Fibroblast Growth Factor/metabolism , Proto-Oncogene Proteins c-akt/metabolism , STAT3 Transcription Factor/metabolism , Carcinoma, Squamous Cell/genetics , Skin Neoplasms/etiology , Skin Neoplasms/prevention & control , Skin Neoplasms/pathology , Ultraviolet Rays/adverse effects , TOR Serine-Threonine Kinases/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Carcinogenesis , RNA, Messenger
2.
Mol Carcinog ; 60(5): 342-353, 2021 05.
Article in English | MEDLINE | ID: mdl-33713497

ABSTRACT

The transcription factor Twist1 has been reported to be essential for the formation and invasiveness of chemically induced tumors in mouse skin. However, the impact of keratinocyte-specific Twist1 deletion on skin carcinogenesis caused by UVB radiation has not been reported. Deletion of Twist1 in basal keratinocytes of mouse epidermis using K5.Cre × Twist1flox/flox mice led to significantly reduced UVB-induced epidermal hyperproliferation. In addition, keratinocyte-specific deletion of Twist1 significantly suppressed UVB-induced skin carcinogenesis. Further analyses revealed that deletion of Twist1 in cultured keratinocytes or mouse epidermis in vivo led to keratinocyte differentiation. In this regard, deletion of Twist1 in epidermal keratinocytes showed significant induction of early and late differentiation markers, including TG1, K1, OVOL1, loricrin, and filaggrin. Similar results were obtained with topical application of harmine, a Harmala alkaloid that leads to degradation of Twist1. In contrast, overexpression of Twist1 in cultured keratinocytes suppressed calcium-induced differentiation. Further analyses using both K5.Cre × Twist1flox/flox mice and an inducible system where Twist1 was deleted in bulge region keratinocytes showed loss of expression of hair follicle stem/progenitor markers, including CD34, Lrig1, Lgr5, and Lgr6. These data support the conclusion that Twist1 has a direct role in maintaining the balance between proliferation and differentiation of keratinocytes and keratinocyte stem/progenitor populations. Collectively, these results demonstrate a critical role for Twist1 early in the process of UVB skin carcinogenesis, and that Twist1 may be a novel target for the prevention of cutaneous squamous cell carcinoma.


Subject(s)
Carcinoma, Squamous Cell/genetics , Skin Neoplasms/genetics , Twist-Related Protein 1/genetics , Ultraviolet Rays/adverse effects , Administration, Topical , Animals , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/etiology , Carcinoma, Squamous Cell/metabolism , Cell Differentiation , Cells, Cultured , Epithelial-Mesenchymal Transition/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockout Techniques , Harmine/administration & dosage , Harmine/pharmacology , Keratinocytes/cytology , Keratinocytes/metabolism , Keratinocytes/pathology , Male , Mice , Skin Neoplasms/drug therapy , Skin Neoplasms/etiology , Skin Neoplasms/metabolism
3.
Mol Carcinog ; 58(9): 1656-1669, 2019 09.
Article in English | MEDLINE | ID: mdl-31237385

ABSTRACT

In this study, we evaluated the role of signal transducer and activator of transcription 1 (STAT1) in response to acute solar ultraviolet (SUV) radiation in mouse epidermis. Analysis of the epidermis from SUV-irradiated mice revealed rapid phosphorylation of STAT1 (pSTAT1) on both tyrosine (tyr701) and serine (ser727) residues and increased levels of IRF-1 while later timepoints showed increased levels of unphosphorylated STAT1 (uSTAT1). STAT1 activation led to upregulation of several proinflammatory chemokine mRNAs in epidermis including Cxcl9, Cxcl10, and Ccl2, as well as, the immune checkpoint inhibitor Pd-l1. In addition, mRNA and protein levels of cyclooxygenase-2 (Cox-2/COX2) were upregulated in epidermis following exposure to SUV. Mice with keratinocyte-specific STAT1 deletion did not exhibit increased IRF-1 or proinflammatory gene expression in epidermis. Furthermore, epidermal COX-2 induction after SUV exposure was significantly reduced in mice with keratinocyte-specific deletion of STAT1. Additionally, SUV irradiation rapidly upregulated interferon gamma (IFNγ) mRNA in the epidermis and that skin resident epidermal CD3 + T-cells were the source of IFNγ production. IFNγ receptor-deficient mice confirmed dependency of STAT1 activation, proinflammatory gene expression and COX-2 upregulation in the epidermis on paracrine IFNγ signaling. Furthermore, keratinocyte-specific STAT1-deficiency reduced proliferation and hyperplasia due to SUV irradiation and this was associated with decreased immune infiltration of mast cells in the dermis. Collectively, the current results demonstrate that exposure to SUV leads to upregulation of IFNγ and downstream pSTAT1/IRF-1/uSTAT1 signaling in the epidermis. Further study of this pathway could lead to identification of novel targets for the prevention of nonmelanoma skin cancer.


Subject(s)
Interferon Regulatory Factor-1/metabolism , Interferon-gamma/metabolism , Keratinocytes/metabolism , STAT1 Transcription Factor/metabolism , Signal Transduction/physiology , Ultraviolet Rays/adverse effects , Animals , CD3 Complex/metabolism , Cyclooxygenase 2/metabolism , Epidermal Cells/metabolism , Epidermis/metabolism , Gene Expression/physiology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , RNA, Messenger/metabolism , Skin/metabolism , Skin Neoplasms/metabolism , T-Lymphocytes/metabolism , Up-Regulation/physiology
4.
Cancer Prev Res (Phila) ; 8(9): 817-25, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26100520

ABSTRACT

In this study, the effects of combining ursolic acid + resveratrol, for possible combined inhibitory effects on skin tumor promotion, were evaluated. Ursolic acid, resveratrol, and the combination of ursolic acid + resveratrol were applied topically prior to 12-O-tetracanoylphorbol-13-acetate (TPA) treatment on mouse skin to examine their effect on TPA-induced signaling pathways, epidermal hyperproliferation, skin inflammation, inflammatory gene expression, and skin tumor promotion. The combination of ursolic acid + resveratrol produced a greater inhibition of TPA-induced epidermal hyperproliferation. The combination of ursolic acid + resveratrol inhibited TPA-induced signaling pathways, including EGFR, STAT3, Src, Akt, Cox-2, Fas, NF-κB, p38 MAPK, c-Jun, and JNK1/2 while increasing levels of tumor suppressors, such as p21 and PDCD4, to a greater extent compared with the groups treated with the individual compounds. Ursolic acid + resveratrol also induced a dramatic increase of p-AMPK-α(Thr172). Combined treatment with ursolic acid + resveratrol resulted in a greater inhibition of expression of proinflammatory cytokines, including Il1a, Il1b, and Il22. Furthermore, NF-κB, Egr-1, and AP-1 DNA binding activities after TPA treatment were dramatically decreased by the combination of ursolic acid + resveratrol. Treatment with ursolic acid + resveratrol during skin tumor promotion with TPA produced greater inhibition of tumor multiplicity and tumor size than with either agent alone. Collectively, the greater ability of the combination of ursolic acid + resveratrol to inhibit skin tumor promotion was due to the greater inhibitory effects on growth factor and inflammatory signaling, skin inflammation, and epidermal hyperproliferation induced by TPA treatment.


Subject(s)
Anticarcinogenic Agents/administration & dosage , Skin Neoplasms/chemically induced , Skin Neoplasms/drug therapy , Stilbenes/administration & dosage , Tetradecanoylphorbol Acetate/chemistry , Triterpenes/administration & dosage , Animals , Carcinogenesis , Cell Nucleus/metabolism , Cell Proliferation , Cytosol/metabolism , Female , Inflammation , Male , Mice , Mice, Inbred ICR , Protein Binding , Resveratrol , Signal Transduction/drug effects , Skin/pathology , Skin Neoplasms/metabolism , Ursolic Acid
5.
Mol Carcinog ; 54(2): 121-33, 2015 Feb.
Article in English | MEDLINE | ID: mdl-24038534

ABSTRACT

STATs play crucial roles in a wide variety of biological functions, including development, proliferation, differentiation and migration as well as in cancer development. In the present study, we examined the impact of constitutive activation of Stat3 on behavior of keratinocytes, including keratinocyte stem cells (KSC) in vivo. BK5.Stat3C transgenic (Tg) mice, which express a constitutively active form of Stat3 (Stat3C) in the basal layer of the epidermis and in the bulge region KSCs exhibited a significantly reduced number of CD34+/α6 integrin+ cells compared to non-transgenic (NTg) littermates. There was a concomitant increase in the Lgr-6, Lrig-1, and Sca-1 populations in the Tg mice in contrast to the CD34 and Keratin-15 positive population. In addition, increased expression of c-myc, ß-catenin, and epithelial-mesenchymal transition (EMT)-related genes as well as decreased expression of α6-integrin was observed in the hair follicles of Tg mice. Notably, Sca-1 was found to be a direct transcriptional target of Stat3 in keratinocytes. The current data suggest that elevated Stat3 activity leads to depletion of hair follicle KSCs along with a concomitant increase of stem/progenitor cells above the bulge region. Overall, the current data indicate that Stat3 plays an important role in keratinocyte stem/progenitor cell homeostasis.


Subject(s)
Endothelial Progenitor Cells/physiology , Keratinocytes/cytology , Keratinocytes/physiology , STAT3 Transcription Factor/metabolism , Animals , Antigens, CD34/genetics , Antigens, CD34/metabolism , Antigens, Ly/genetics , Antigens, Ly/metabolism , Antigens, Surface/genetics , Antigens, Surface/metabolism , Bromodeoxyuridine/pharmacology , Cell Differentiation , Cell Line , Epithelial-Mesenchymal Transition , Female , Gene Expression Regulation , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Transgenic , NIH 3T3 Cells , STAT3 Transcription Factor/genetics
6.
J Invest Dermatol ; 134(7): 1971-1980, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24496235

ABSTRACT

The nuclear transcription factor signal transducer and activator of transcription 3 (Stat3) has recently been reported to have a localized mitochondrial regulatory function. Current data suggest that mitochondrial Stat3 (mitoStat3) is necessary for maximal mitochondrial activity and for Ras-mediated transformation independent of Stat3 nuclear activity. We have previously shown that Stat3 has a pivotal role in epithelial carcinogenesis. Therefore, the aim of the current study was to determine the role of mitoStat3 in epidermal keratinocytes. Herein, we show that normal and neoplastic keratinocytes contain a pool of mitoStat3. EGF and 12-O-tetradecanoylphorbol-13-acetate induce Stat3 mitochondrial translocation mediated through the phosphorylation of Stat3 at Ser727. In addition, we report that mitoStat3 binds mtDNA and associates with the mitochondrial transcription factor A. Furthermore, Stat3 ablation resulted in an increase of mitochondrial-encoded gene transcripts. An increase in key nuclear-encoded metabolic genes, PGC-1α and NRF-1, was also observed in Stat3-null keratinocytes; however, no changes in nuclear-encoded electron transport chain gene transcripts or mtDNA copy number were observed. Collectively, our findings suggest a heretofore-unreported function for mitoStat3 as a potential mitochondrial transcription factor in keratinocytes. This mitoStat3-mtDNA interaction may represent an alternate signaling pathway that could alter mitochondrial function and biogenesis and have a role in tumorigenesis.


Subject(s)
Cell Transformation, Neoplastic/metabolism , DNA, Mitochondrial/metabolism , Keratinocytes/physiology , STAT3 Transcription Factor/metabolism , Skin Neoplasms/metabolism , Animals , Cell Transformation, Neoplastic/genetics , Epidermal Cells , Epidermis/physiology , Gene Expression/physiology , Humans , Keratinocytes/cytology , Mice , Mitochondria/genetics , Mitochondria/metabolism , Primary Cell Culture , Signal Transduction/physiology , Skin Neoplasms/genetics
7.
Proc Natl Acad Sci U S A ; 110(8): E687-96, 2013 Feb 19.
Article in English | MEDLINE | ID: mdl-23386725

ABSTRACT

DNA polymerase ζ (polζ) is critical for bypass of DNA damage and the associated mutagenesis, but also has unique functions in mammals. It is required for embryonic development and for viability of hematopoietic cells, but, paradoxically, skin epithelia appear to survive polζ deletion. We wished to determine whether polζ functions in a tissue-specific manner and how polζ status influences skin tumorigenesis. Mice were produced in which Rev3L (the catalytic subunit of polζ) was deleted in tissues expressing keratin 5. Efficient epidermal deletion of Rev3L was tolerated but led to skin and hair abnormalities, accompanied by evidence of DNA breaks. Unchallenged mice developed tumors in keratin 5-expressing tissues with age, consistent with the chromosomal instability accompanying a polζ defect. Unexpectedly, mice with the Rev3L deletion were much more sensitive to UVB radiation than mice defective in other DNA repair genes. Following irradiation, polζ-defective mice failed to mount skin-regenerative responses and responded to stress by mobilizing melanocytes to the epidermis. However, they did not develop skin tumors after chronic UVB irradiation. To determine the proliferative potential of polζ-deficient skin epithelia, keratinocytes were isolated and examined. These keratinocytes harbored chromosomal gaps and breaks and exhibited a striking proliferation defect. These results can be unified by a model in which slowly dividing cells accumulate replication-associated DNA breaks but otherwise survive Rev3L deletion, but functional polζ is essential for responses requiring rapid proliferation, both in cell culture and in vivo. The results reveal a biological role for mammalian polζ in tolerating DNA damage and enabling proliferative responses in vivo.


Subject(s)
Cell Proliferation , Genomic Instability , Animals , Carcinoma, Squamous Cell/genetics , DNA-Binding Proteins/genetics , DNA-Directed DNA Polymerase/genetics , DNA-Directed DNA Polymerase/metabolism , Gene Deletion , Mice , Skin Neoplasms/genetics , Ultraviolet Rays
8.
Cancer Prev Res (Phila) ; 5(10): 1236-46, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22896210

ABSTRACT

Obesity, an established risk factor for epithelial cancers, remains prevalent in the United States and many other countries. In contrast to positive energy balance states (overweight, obesity), calorie restriction (CR) has been shown to act as a universal inhibitor of tumorigenesis in multiple animal models of human cancer. Unfortunately, the mechanisms underlying the enhancing effects of obesity or the inhibitory effects of CR on cancer etiology remain elusive. Here, we evaluated the impact of dietary energy balance manipulation on epithelial carcinogenesis and identified several potential mechanisms that may account for the differential effects of obesity and CR on cancer. Obesity enhanced tumor promotion during epithelial carcinogenesis, in part, due to altered insulin-like growth factor-1 receptor (IGF-1R)/EGF receptor (EGFR) crosstalk and downstream signaling to effectors such as Akt/mTOR. Obesity-induced changes in cellular signaling subsequently led to altered levels of cell-cycle proteins that favored enhanced epidermal proliferation during tumor promotion. In contrast, CR reduced susceptibility to tumor promotion, attenuated IGF-1R/EGFR crosstalk and downstream signaling, and altered levels of cell-cycle proteins that favored reduced epidermal proliferation during tumor promotion. Collectively, these findings suggest potential targets for the prevention of epithelial cancers, as well as for reversal of obesity-mediated cancer development and progression. Cancer Prev Res; 5(10); 1236-46. ©2012 AACR.


Subject(s)
Cell Transformation, Neoplastic/pathology , Energy Metabolism , ErbB Receptors/metabolism , Papilloma/pathology , Receptor, IGF Type 1/metabolism , Skin Neoplasms/pathology , 9,10-Dimethyl-1,2-benzanthracene/toxicity , Animals , Blotting, Western , Caloric Restriction , Carcinogens/toxicity , Cell Cycle , Cell Proliferation , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/metabolism , Cells, Cultured , Diet , ErbB Receptors/genetics , Female , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/metabolism , Keratinocytes/cytology , Keratinocytes/drug effects , Keratinocytes/metabolism , Mice , Mice, Inbred ICR , Obesity , Papilloma/chemically induced , Papilloma/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Receptor, IGF Type 1/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Skin Neoplasms/chemically induced , Skin Neoplasms/metabolism , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism
9.
PLoS One ; 6(4): e18266, 2011 Apr 04.
Article in English | MEDLINE | ID: mdl-21483750

ABSTRACT

Development of a suitable mouse model would facilitate the investigation of pathomechanisms underlying human psoriasis and would also assist in development of therapeutic treatments. However, while many psoriasis mouse models have been proposed, no single model recapitulates all features of the human disease, and standardized validation criteria for psoriasis mouse models have not been widely applied. In this study, whole-genome transcriptional profiling is used to compare gene expression patterns manifested by human psoriatic skin lesions with those that occur in five psoriasis mouse models (K5-Tie2, imiquimod, K14-AREG, K5-Stat3C and K5-TGFbeta1). While the cutaneous gene expression profiles associated with each mouse phenotype exhibited statistically significant similarity to the expression profile of psoriasis in humans, each model displayed distinctive sets of similarities and differences in comparison to human psoriasis. For all five models, correspondence to the human disease was strong with respect to genes involved in epidermal development and keratinization. Immune and inflammation-associated gene expression, in contrast, was more variable between models as compared to the human disease. These findings support the value of all five models as research tools, each with identifiable areas of convergence to and divergence from the human disease. Additionally, the approach used in this paper provides an objective and quantitative method for evaluation of proposed mouse models of psoriasis, which can be strategically applied in future studies to score strengths of mouse phenotypes relative to specific aspects of human psoriasis.


Subject(s)
Gene Expression Profiling/methods , Genomics/methods , Psoriasis/genetics , Adolescent , Adult , Aged , Aminoquinolines/therapeutic use , Animals , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Epidermis/metabolism , Humans , Imiquimod , Keratinocytes/metabolism , Leukocytes/immunology , Mice , Middle Aged , Mitosis/genetics , Phenotype , Psoriasis/immunology , Psoriasis/metabolism , Psoriasis/pathology , Receptor Protein-Tyrosine Kinases/genetics , Receptor, TIE-2 , Transcription, Genetic , Young Adult
10.
Neoplasia ; 13(3): 254-65, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21390188

ABSTRACT

In this report, we describe the development of a transgenic mouse in which a rat probasin promoter (ARR(2)Pb) was used to direct prostate specific expression of a constitutively active form of signal transducer and activator of transcription 3 (i.e., Stat3C). ARR(2)Pb.Stat3C mice exhibited hyperplasia and prostate intraepithelial neoplasia (PIN) lesions in both ventral and dorsolateral prostate lobes at 6 and 12 months; however, no adenocarcinomas were detected. The effect of combined loss of PTEN was examined by crossing ARR(2)Pb.Stat3C mice with PTEN(+/-) null mice. PTEN(+/-) null mice on an ICR genetic background developed only hyperplasia and PIN at 6 and 12 months, respectively. ARR(2)Pb.Stat3C x PTEN(+/-) mice exhibited a more severe prostate phenotype compared with ARR(2)Pb.Stat3C and PTEN(+/-) mice. ARR(2)Pb.Stat3C x PTEN(+/-) mice developed adenocarcinomas in the ventral prostate as early as 6 months (22% incidence) that reached an incidence of 61% by 12 months. Further evaluations indicated that phospho-Stat3, phospho-Akt, phospho-nuclear factor κB, cyclin D1, and Ki67 were upregulated in adenocarcinomas from ARR(2)Pb.Stat3C x PTEN(+/-) mice. In addition, membrane staining for ß-catenin and E-cadherin was reduced. The changes in Stat3 and nuclear factor κB phosphorylation correlated most closely with tumor progression. Collectively, these data provide evidence that Stat3 and Akt signaling cooperate in prostate cancer development and progression and that ARR(2)Pb.Stat3C x PTEN(+/-) mice represent a novel mouse model of prostate cancer to study these interactions.


Subject(s)
Adenocarcinoma/pathology , Prostatic Intraepithelial Neoplasia/pathology , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins c-akt/physiology , STAT3 Transcription Factor/physiology , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Androgen-Binding Protein/genetics , Animals , Blotting, Western , Disease Progression , Female , Humans , Male , Mice , Mice, Knockout , Mice, Transgenic , PTEN Phosphohydrolase/physiology , Phosphorylation , Polymerase Chain Reaction , Prostate/metabolism , Prostate/pathology , Prostatic Intraepithelial Neoplasia/genetics , Prostatic Intraepithelial Neoplasia/metabolism , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Rats , Signal Transduction
11.
J Natl Cancer Inst ; 100(24): 1815-7, 2008 Dec 17.
Article in English | MEDLINE | ID: mdl-19066276

ABSTRACT

Noncanonical DNA structures are postulated to be responsible for some breakpoint hotspots that occur frequently in cancers. We developed a novel mouse model system using the naturally occurring H-DNA structure that deviate from the familiar right-handed helical B form found at the breakage hotspot in the human c-MYC promoter and a Z-DNA-forming CG repeat to test this idea directly. Large-scale chromosomal deletions and/or translocations occurred in 5 (7.7%, 95% confidence interval [CI] = 3.7% to 12.8%) of the 65 mice carrying the H-DNA-forming sequences and in 7 (6.6%, 95% CI = 3.8% to 11.6%) of the 106 mice carrying the Z-DNA-forming sequences, but in 0 of the 63 control mice (P = .042 and P = .035, respectively, two-sided test). Thus, the DNA structure itself can introduce instability in a mammalian genome.


Subject(s)
Chromosome Aberrations , DNA, Neoplasm , DNA, Z-Form , DNA , Genomic Instability , Animals , Chromosome Breakage , Chromosome Deletion , Chromosome Inversion , Disease Models, Animal , Gene Expression Regulation, Neoplastic , Mice , Mice, Transgenic , Mutation , Polymerase Chain Reaction , Translocation, Genetic
12.
Cancer Cell ; 14(3): 212-25, 2008 Sep 09.
Article in English | MEDLINE | ID: mdl-18772111

ABSTRACT

It has long been known that excessive mitotic activity due to H-Ras can block keratinocyte differentiation and cause skin cancer. It is not clear whether there are any innate surveillants that are able to ensure that keratinocytes undergo terminal differentiation, preventing the disease. IKKalpha induces keratinocyte terminal differentiation, and its downregulation promotes skin tumor development. However, its intrinsic function in skin cancer is unknown. Here, we found that mice with IKKalpha deletion in keratinocytes develop a thickened epidermis and spontaneous squamous cell-like carcinomas. Inactivation of epidermal growth factor receptor (EGFR) or reintroduction of IKKalpha inhibits excessive mitosis, induces terminal differentiation, and prevents skin cancer through repressing an EGFR-driven autocrine loop. Thus, IKKalpha serves as an innate surveillant.


Subject(s)
Homeostasis , I-kappa B Kinase/metabolism , Skin Neoplasms/metabolism , Skin/metabolism , ADAM Proteins/genetics , ADAM Proteins/metabolism , Amphiregulin , Animals , Animals, Newborn , EGF Family of Proteins , Epidermal Growth Factor/genetics , Epidermis/metabolism , Epidermis/pathology , ErbB Receptors/genetics , ErbB Receptors/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Gene Expression Regulation , Glycoproteins/genetics , I-kappa B Kinase/genetics , Intercellular Signaling Peptides and Proteins/genetics , Keratinocytes/metabolism , Keratinocytes/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Knockout , Mice, Transgenic , Promoter Regions, Genetic/genetics , Protein Binding/drug effects , Proto-Oncogene Proteins p21(ras)/metabolism , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor/metabolism , Skin/pathology , Skin Neoplasms/pathology
13.
Cancer Res ; 68(10): 3680-8, 2008 May 15.
Article in English | MEDLINE | ID: mdl-18483250

ABSTRACT

Calorie restriction has been shown to inhibit epithelial carcinogenesis and this method of dietary restriction reduces many circulating proteins, including insulin-like growth factor I (IGF-I). Previously, we identified a relationship between elevated tissue IGF-I levels and enhanced susceptibility to chemically induced skin tumorigenesis. In this study, liver IGF-I-deficient (LID) mice, which have a 75% reduction in serum IGF-I, were subjected to the standard two-stage skin carcinogenesis protocol using 7,12-dimethylbenz(a)anthracene as the initiator and 12-O-tetradecanoylphorbol-13-acetate (TPA) as the promoter. We observed a significant reduction in epidermal thickness and labeling index in LID mice treated with either vehicle or TPA. A significant decrease in both tumor incidence and tumor multiplicity was observed in LID mice undergoing two-stage skin carcinogenesis relative to wild-type littermates. Western blot analyses of epidermal extracts revealed reduced activation of both the epidermal growth factor and IGF-I receptors in response to TPA treatment in LID mice. In addition, reduced activation of both Akt and the mammalian target of rapamycin (mTOR) was observed in LID mice following TPA treatment relative to wild-type controls. Signaling downstream of mTOR was also reduced. These data suggest a possible mechanism whereby reduced circulating IGF-I leads to attenuated activation of the Akt and mTOR signaling pathways, and thus, diminished epidermal response to tumor promotion, and ultimately, two-stage skin carcinogenesis. The current data also suggest that reduced circulating IGF-I levels which occur as a result of calorie restriction may lead to the inhibition of skin tumorigenesis, at least in part, by a similar mechanism.


Subject(s)
Genetic Predisposition to Disease , Insulin-Like Growth Factor I/biosynthesis , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Animals , Benz(a)Anthracenes/pharmacology , Cell Proliferation , Cell Transformation, Neoplastic , Epidermis/metabolism , Female , Mice , Models, Biological , Neoplasms/metabolism , Protein Kinases/metabolism , Signal Transduction , Skin Neoplasms/chemically induced , TOR Serine-Threonine Kinases , Tetradecanoylphorbol Acetate/pharmacology
14.
Carcinogenesis ; 29(6): 1108-14, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18453544

ABSTRACT

Constitutive activation of signal transducer and activator of transcription 3 (Stat3) has been found in a variety of human malignancies and has been suggested to play an important role in carcinogenesis. Recently, our laboratory demonstrated that Stat3 is required for the development of skin tumors via two-stage carcinogenesis using skin-specific loss-of-function transgenic mice. To investigate further the role of Stat3 in each stage of chemical carcinogenesis in mouse skin, i.e. initiation and promotion stages, we generated inducible Stat3-deficient mice (K5.Cre-ER(T2) x Stat3(fl/fl)) that show epidermal-specific disruption of Stat3 following topical treatment with 4-hydroxytamoxifen (TM). The epidermis of inducible Stat3-deficient mice treated with TM showed a significant increase in apoptosis induced by 7,12-dimethylbenz[a]anthracene (DMBA) and reduced proliferation following exposure to 12-O-tetradecanoylphorbol-13-acetate. In two-stage skin carcinogenesis assays, inducible Stat3-deficient mice treated with TM during the promotion stage showed a significant delay of tumor development and a significantly reduced number of tumors compared with control groups. Inducible Stat3-deficient mice treated with TM before initiation with DMBA also showed a significant delay in tumor development and a significantly reduced number of tumors compared with control groups. Finally, treatment of inducible Stat3-deficient mice that had existing skin tumors generated by the two-stage carcinogenesis protocol with TM (by intraperitoneal injection) led to inhibition of tumor growth compared with tumors formed in control groups. Collectively, these results directly demonstrate that Stat3 is required for skin tumor development during both the initiation and promotion stages of skin carcinogenesis in vivo.


Subject(s)
Cell Transformation, Neoplastic/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction/physiology , Skin Neoplasms/metabolism , 9,10-Dimethyl-1,2-benzanthracene/toxicity , Animals , Blotting, Western , Carcinogens/toxicity , Immunohistochemistry , Mice , Mice, Transgenic , Polymerase Chain Reaction , STAT3 Transcription Factor/genetics , Skin Neoplasms/chemically induced , Skin Neoplasms/genetics , Tamoxifen/analogs & derivatives , Tamoxifen/toxicity , Tetradecanoylphorbol Acetate/toxicity
15.
Cancer Prev Res (Phila) ; 1(1): 65-76, 2008 Jun.
Article in English | MEDLINE | ID: mdl-19138937

ABSTRACT

The prevalence of obesity, an established risk factor for several types of cancer, has increased steadily over the past several decades in the United States. New targets and strategies for offsetting the effect of obesity on cancer risk are urgently needed. In the present study, we examined the effect of dietary energy balance manipulation on steady-state signaling in multiple epithelial tissues, with a focus on the Akt and mammalian target of rapamycin (mTOR) pathways. For these experiments, male FVB/N and C57BL/6 and female ICR mice were maintained on a control (10 kcal% fat) diet, a diet-induced obesity (DIO; 60 kcal% fat) regimen, or a 30% calorie restriction (CR) regimen for 15 to 17 weeks. Relative to the control group, the DIO regimen increased, whereas CR decreased, circulating insulin-like growth factor-I (IGF-I) as has previously been reported. Western blot analyses showed that the DIO regimen enhanced, whereas CR inhibited, activation of Akt and mTOR, regardless of epithelial tissue or genetic background. In contrast, activation of AMP-activated protein kinase was modulated by dietary energy balance manipulation in the liver but not in the epidermis or dorsolateral prostate. Western blot analyses of epidermal extracts taken from ICR mice also revealed reduced activation of both the IGF-I receptor and epidermal growth factor receptor in CR mice, compared with control mice or mice maintained on the DIO regimen. Taken together, these novel findings suggest that dietary energy balance modulates signaling through cell-surface receptors (i.e., IGF-I receptor and epidermal growth factor receptor), affecting activation of multiple downstream pathways including Akt and mTOR, thus providing important dietary and pharmacologic targets for disrupting the obesity-cancer link.


Subject(s)
Diet , Energy Metabolism/physiology , Epithelium/metabolism , Oncogene Protein v-akt/physiology , Protein Kinases/physiology , Animals , Body Fat Distribution , Body Weight , Eating/physiology , Female , Homeostasis/physiology , Insulin-Like Growth Factor I/analysis , Insulin-Like Growth Factor I/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Oncogene Protein v-akt/metabolism , Protein Kinases/metabolism , Signal Transduction/physiology , TOR Serine-Threonine Kinases
16.
Mol Biol Cell ; 19(1): 137-49, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17959825

ABSTRACT

Aberrant activation of the Akt pathway has been implicated in several human pathologies including cancer. However, current knowledge on the involvement of Akt signaling in development is limited. Previous data have suggested that Akt-mediated signaling may be an essential mediator of epidermal homeostasis through cell autonomous and noncell autonomous mechanisms. Here we report the developmental consequences of deregulated Akt activity in the basal layer of stratified epithelia, mediated by the expression of a constitutively active Akt1 (myrAkt) in transgenic mice. Contrary to mice overexpressing wild-type Akt1 (Akt(wt)), these myrAkt mice display, in a dose-dependent manner, altered development of ectodermally derived organs such as hair, teeth, nails, and epidermal glands. To identify the possible molecular mechanisms underlying these alterations, gene profiling approaches were used. We demonstrate that constitutive Akt activity disturbs the bone morphogenetic protein-dependent signaling pathway. In addition, these mice also display alterations in adult epidermal stem cells. Collectively, we show that epithelial tissue development and homeostasis is dependent on proper regulation of Akt expression and activity.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Ectoderm/abnormalities , Ectoderm/enzymology , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Animals , Ectoderm/pathology , Enzyme Activation , Epidermis/enzymology , Epidermis/pathology , Forkhead Box Protein O3 , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Gene Expression Regulation, Enzymologic , Hair/abnormalities , Hair/ultrastructure , Homeostasis , Mice , Mice, Transgenic , Nails, Malformed/enzymology , Oligonucleotide Array Sequence Analysis , Phenotype , Proto-Oncogene Proteins c-akt/genetics , Stem Cells/cytology , Stem Cells/enzymology , Tooth Abnormalities/enzymology
17.
Cancer Res ; 67(22): 10879-88, 2007 Nov 15.
Article in English | MEDLINE | ID: mdl-18006833

ABSTRACT

Aberrant activation of the phosphoinositide-3-kinase (PI3K)/PTEN/Akt pathway, leading to increased proliferation and decreased apoptosis, has been implicated in several human pathologies including cancer. Our previous data have shown that Akt-mediated signaling is an essential mediator in the mouse skin carcinogenesis system during both the tumor promotion and progression stages. In addition, overexpression of Akt is also able to transform keratinocytes through transcriptional and posttranscriptional processes. Here, we report the consequences of the increased expression of Akt1 (wtAkt) or constitutively active Akt1 (myrAkt) in the basal layer of stratified epithelia using the bovine keratin K5 promoter. These mice display alterations in epidermal proliferation and differentiation. In addition, transgenic mice with the highest levels of Akt expression developed spontaneous epithelial tumors in multiple organs with age. Furthermore, both wtAkt and myrAkt transgenic lines displayed heightened sensitivity to the epidermal proliferative effects of the tumor promoter 12-O-tetradecanoylphorbol-13-acetate (TPA) and heightened sensitivity to two-stage skin carcinogenesis. Finally, enhanced susceptibility to two-stage carcinogenesis correlated with a more sustained proliferative response following treatment with TPA as well as sustained alterations in Akt downstream signaling pathways and elevations in cell cycle regulatory proteins. Collectively, the data provide direct support for an important role for Akt signaling in epithelial carcinogenesis in vivo, especially during the tumor promotion stage.


Subject(s)
Epithelium/pathology , Gene Expression Regulation, Neoplastic , Gene Expression Regulation , Proto-Oncogene Proteins c-akt/metabolism , Skin Neoplasms/chemically induced , Skin Neoplasms/pathology , Animals , Cattle , Epithelium/metabolism , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Fluorescence , Promoter Regions, Genetic , Signal Transduction , Skin Neoplasms/metabolism , Tetradecanoylphorbol Acetate/toxicity
18.
Cancer Res ; 67(8): 3794-800, 2007 Apr 15.
Article in English | MEDLINE | ID: mdl-17440093

ABSTRACT

The macrolide fungicide rapamycin has shown significant antiproliferative action toward a variety of tumor types. In this study, we used BK5.erbB2 transgenic mice as an animal model to examine the therapeutic effect of rapamycin as a potential treatment for gallbladder cancer. Homozygous BK5.erbB2 mice overexpressing the wild-type rat erbB2 gene in basal epithelial cells of the gallbladder have an approximately 70% incidence of gallbladder adenocarcinoma by 2 to 3 months of age. Groups of mice ( approximately 2-3 months of age) were treated with rapamycin by i.p. injection (once daily for 14 days) and then sacrificed 24 h after the last treatment. Rapamycin significantly reduced the incidence and severity of gallbladder carcinoma in BK5.erbB2 mice in a dose-dependent manner. Tumors responsive to treatment exhibited a higher number of apoptotic cells. Furthermore, rapamycin treatment led to decreased levels of phosphorylated p70 S6 kinase (Thr(389)) in gallbladder tissue as assessed by both Western blot and immunofluorescence analyses. Finally, immunofluorescence staining revealed elevated phosphorylated Akt (Ser(473)) and phosphorylated mammalian target of rapamycin (mTOR; Ser(2448)) in human gallbladder cancer compared with normal gallbladder tissue. Based on our results using a novel genetically engineered mouse model and the fact that the Akt/mTOR pathway is activated in human gallbladder cancer, rapamycin and related drugs may be effective therapeutic agents for the treatment of human gallbladder cancer.


Subject(s)
Adenocarcinoma/drug therapy , Antibiotics, Antineoplastic/pharmacology , Gallbladder Neoplasms/drug therapy , Sirolimus/pharmacology , Adenocarcinoma/metabolism , Animals , Female , Gallbladder Neoplasms/metabolism , Humans , Male , Mice , Mice, Inbred ICR , Mice, Transgenic , Phosphorylation/drug effects , Protein Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , TOR Serine-Threonine Kinases
19.
Mol Carcinog ; 44(2): 137-45, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16086373

ABSTRACT

Overexpression of human IGF-1 with the bovine keratin 5 (BK5) promoter (BK5.IGF-1 transgenic mice) induces persistent epidermal hyperplasia and leads to spontaneous skin tumor formation. In previous work, PI3K and Akt activities were found to be elevated in the epidermis of BK5.IGF-1 transgenic mice compared to nontransgenic littermates. In the present study, we examined the importance of the PI3K/Akt signaling pathway in mediating the skin phenotype and the skin tumor promoting action of IGF-1 in these mice. Western blot analyses with epidermal lysates showed that signaling components downstream of PI3K/Akt were altered in epidermis of BK5.IGF-1 mice. Increased phosphorylation of GSK-3 (Ser(9/21)), TSC2(Thr(1462)), and mTOR(Ser(2448)) was observed. In addition, hypophosphorylation and increased protein levels of beta-catenin were observed in the epidermis of BK5.IGF-1 mice. These data suggested that components downstream of Akt might be affected, including cell cycle machinery in the epidermis of BK5.IGF-1 mice. Protein levels of cyclins (D1, E, A), E2F1, and E2F4 were all elevated in the epidermis of BK5.IGF-1 mice. Also, immunoprecipitation experiments demonstrated an increase in cdk4/cyclin D1 and cdk2/cyclin E complex formation, suggesting increased cdk activity in the epidermis of transgenic mice. In further studies, the PI3K inhibitor, LY294002, significantly blocked IGF-1-mediated epidermal proliferation and skin tumor promotion in DMBA-initiated BK5.IGF-1 mice. In addition, inhibition of PI3K/Akt with LY294002 reversed many of the cell cycle related changes observed in untreated transgenic animals. Collectively, the current results supported the hypothesis that elevated PI3K/Akt activity and subsequent activation of one or more downstream effector pathways contributed significantly to the tumor promoting action of IGF-1 in the epidermis of BK5.IGF-1 mice.


Subject(s)
Insulin-Like Growth Factor I/physiology , Phosphatidylinositol 3-Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Signal Transduction , Skin Neoplasms/metabolism , Animals , Cell Cycle Proteins/metabolism , Chromones/pharmacology , Epidermis/metabolism , Female , Insulin-Like Growth Factor I/pharmacology , Mice , Mice, Transgenic , Morpholines/pharmacology , Proto-Oncogene Proteins c-akt
20.
Nat Med ; 11(1): 43-9, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15592573

ABSTRACT

Here we report that epidermal keratinocytes in psoriatic lesions are characterized by activated Stat3. Transgenic mice with keratinocytes expressing a constitutively active Stat3 (K5.Stat3C mice) develop a skin phenotype either spontaneously, or in response to wounding, that closely resembles psoriasis. Keratinocytes from K5.Stat3C mice show upregulation of several molecules linked to the pathogenesis of psoriasis. In addition, the development of psoriatic lesions in K5.Stat3C mice requires cooperation between Stat3 activation in keratinocytes and activated T cells. Finally, abrogation of Stat3 function by a decoy oligonucleotide inhibits the onset and reverses established psoriatic lesions in K5.Stat3C mice. Thus, targeting Stat3 may be potentially therapeutic in the treatment of psoriasis.


Subject(s)
DNA-Binding Proteins/metabolism , Epidermis/metabolism , Keratinocytes/metabolism , Psoriasis/metabolism , T-Lymphocytes/metabolism , Trans-Activators/metabolism , Animals , DNA-Binding Proteins/genetics , Disease Models, Animal , Epidermis/immunology , Humans , Keratinocytes/immunology , Mice , Mice, Transgenic , Psoriasis/genetics , Psoriasis/immunology , STAT3 Transcription Factor , Severe Combined Immunodeficiency/metabolism , Signal Transduction/genetics , Signal Transduction/physiology , Skin Transplantation , T-Lymphocytes/immunology , Trans-Activators/genetics , Transplantation, Heterologous
SELECTION OF CITATIONS
SEARCH DETAIL
...