Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Cancer Res ; 71(6): 2318-27, 2011 Mar 15.
Article in English | MEDLINE | ID: mdl-21406403

ABSTRACT

Protein kinase C epsilon (PKCε), a novel PKC isoform, is overexpressed in prostate cancer (PCa) and correlates with disease aggressiveness. However, the functional contribution of PKCε to development or progression of PCa remained to be determined. Here we present the first in vivo genetic evidence that PKCε is essential for both the development and metastasis of PCa in the transgenic mouse model of prostate adenocarcinoma (TRAMP). Heterozygous or homozygous genetic deletions of PKCε in FVB/N TRAMP inhibited PCa development and metastasis as analyzed by positron emission tomography/computed tomography, tumor weight determinations, and histopathology. We also examined biomarkers associated with tumor progression in this model, including markers of survival, proliferation, angiogenesis, inflammation, and metastatic progression. To find clues about the genes regulated by PKCε and linked to the Stat3 signaling pathway, we carried out focused PCR arrays of JAK/STAT signaling in excised PCa tissues from PKCε wild-type and nullizygous TRAMP mice. Notably, PKCε loss was associated with significant downregulation of proliferative and metastatic genes C/EBPß (CCAAT/enhancer binding protein ß), CRP (C-reactive protein), CMK, EGFR (epidermal growth factor receptor), CD64, Jun B, and gp130. Taken together, our findings offer the first genetic evidence of the role of PKCε in PCa development and metastasis. PKCε may be potential target for prevention and/or treatment of PCa.


Subject(s)
Adenocarcinoma/metabolism , Disease Models, Animal , Prostatic Neoplasms/metabolism , Protein Kinase C-epsilon/metabolism , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Animals , C-Reactive Protein/genetics , ErbB Receptors/genetics , Female , Gene Expression , Humans , Immunoblotting , Janus Kinases/metabolism , Male , Mice , Mice, Knockout , Mice, Transgenic , Neoplasm Metastasis , Positron-Emission Tomography , Prostate/metabolism , Prostate/pathology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Protein Kinase C-epsilon/genetics , Reverse Transcriptase Polymerase Chain Reaction , STAT3 Transcription Factor/metabolism , Signal Transduction , Tomography, X-Ray Computed
2.
Cancer Prev Res (Phila) ; 3(1): 35-47, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20051371

ABSTRACT

Preclinical studies have shown that the inhibition of ornithine decarboxylase (ODC) by alpha-difluoromethylornithine (DFMO) and resultant decreases in tissue concentrations of polyamines (putrescine and spermidine) prevents neoplastic developments in many tissue types. Clinical studies of oral DFMO at 500 mg/m(2)/day revealed it to be safe and tolerable and resulted in significant inhibition of phorbol ester-induced skin ODC activity. Two hundred and ninety-one participants (mean age, 61 years; 60% male) with a history of prior nonmelanoma skin cancer (NMSC; mean, 4.5 skin cancers) were randomized to oral DFMO (500 mg/m(2)/day) or placebo for 4 to 5 years. There was a trend toward a history of more prior skin cancers in subjects randomized to placebo, but all other characteristics including sunscreen and nonsteroidal anti-inflammatory drug use were evenly distributed. Evaluation of 1,200 person-years of follow-up revealed a new NMSC rate of 0.5 events/person/year. The primary end point, new NMSCs, was not significantly different between subjects taking DFMO and placebo (260 versus 363 cancers, P = 0.069, two-sample t test). Evaluation of basal cell (BCC) and squamous cell cancers separately revealed very little difference in squamous cell cancer between treatment groups but a significant difference in new BCC (DFMO, 163 cancers; placebo, 243 cancers; expressed as event rate of 0.28 BCC/person/year versus 0.40 BCC/person/year, P = 0.03). Compliance with DFMO was >90% and it seemed to be well tolerated with evidence of mild ototoxicity as measured by serial audiometric examination when compared with placebo subjects. The analysis of normal skin biopsies revealed a significant (P < 0.05) decrease in 12-0-tetradecanoylphorbol-13-acetate-induced ODC activity (month 24, 36, and 48) and putrescine concentration (month 24 and 36 only) in DFMO subjects. Subjects with a history of skin cancer taking daily DFMO had an insignificant reduction (P = 0.069) in new NMSC that was predominantly due to a marked reduction in new BCC. Based on these data, the potential of DFMO, alone or in combination, to prevent skin cancers should be explored further.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Basal Cell/prevention & control , Carcinoma, Squamous Cell/prevention & control , Eflornithine/therapeutic use , Skin Neoplasms/prevention & control , Audiometry , Carcinoma, Basal Cell/mortality , Carcinoma, Squamous Cell/mortality , Double-Blind Method , Female , Hearing/drug effects , Humans , Kaplan-Meier Estimate , Male , Medication Adherence , Middle Aged , Ornithine Decarboxylase Inhibitors , Skin Neoplasms/mortality
3.
J Invest Dermatol ; 130(1): 270-7, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19626035

ABSTRACT

Chronic exposure to UVR is the major etiologic factor in the development of human skin cancers including squamous-cell carcinoma (SCC). We have previously shown that protein Kinase C epsilon (PKCepsilon) transgenic mice on FVB/N background, which overexpress PKCepsilon protein approximately eightfold over endogenous levels in epidermis, exhibit about threefold more sensitivity than wild-type littermates to UVR-induced development of SCC. To determine whether it is PKCepsilon and not the mouse genetic background that determines susceptibility to UVR carcinogenesis, we cross-bred PKCepsilon FVB/N transgenic mice with SKH-1 hairless mice to generate PKCepsilon-overexpressing SKH-1 hairless mice. To evaluate the susceptibility of PKCepsilon SKH-1 hairless transgenic mice to UVR carcinogenesis, the mice were exposed to UVR (1-2 KJ m(-2)) three times weekly from a bank of six kodacel-filtered FS40 sunlamps. As compared with the wild-type hairless mice, PKCepsilon overexpression in SKH-1 hairless mice decreased the latency (12 weeks), whereas it increased the incidence (twofold) and multiplicity (fourfold) of SCC. The SKH hairless transgenic mice were observed to be as sensitive as FVB/N transgenic mice to UVR-induced development of SCC and expression of proliferative markers (proliferating cell nuclear antigen, signal transducers and activators of transcription 3, and extracellular signal-regulated kinase 1/2). The results indicate that PKCepsilon level dictates susceptibility, irrespective of genetic background, to UVR carcinogenesis.


Subject(s)
Carcinoma, Squamous Cell/enzymology , Carcinoma, Squamous Cell/genetics , Protein Kinase C-epsilon/genetics , Skin Neoplasms/enzymology , Skin Neoplasms/genetics , Ultraviolet Rays/adverse effects , Animals , Cell Nucleus/metabolism , Dose-Response Relationship, Radiation , Epidermis/physiology , Epidermis/radiation effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Genetic Predisposition to Disease , Male , Mice , Mice, Hairless , Mice, Transgenic , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/radiation effects , Proliferating Cell Nuclear Antigen/metabolism , Protein Kinase C-epsilon/metabolism , STAT3 Transcription Factor/metabolism , Species Specificity
4.
Cancer Res ; 68(21): 9024-32, 2008 Nov 01.
Article in English | MEDLINE | ID: mdl-18974148

ABSTRACT

Prostate cancer (PCa) is the second leading cause of cancer-related deaths in men. Hormone-refractory invasive PCa is the end stage and accounts for the majority of PCa patient deaths. We present here that plumbagin (PL), a quinoid constituent isolated from the root of the medicinal plant Plumbago zeylanica L., may be a potential novel agent in the control of hormone-refractory PCa. Specific observations are the findings that PL inhibited PCa cell invasion and selectively induced apoptosis in PCa cells but not in immortalized nontumorigenic prostate epithelial RWPE-1 cells. In addition, i.p. administration of PL (2 mg/kg body weight), beginning 3 days after ectopic implantation of hormone-refractory DU145 PCa cells, delayed tumor growth by 3 weeks and reduced both tumor weight and volume by 90%. Discontinuation of PL treatment in PL-treated mice for as long as 4 weeks did not result in progression of tumor growth. PL, at concentrations as low as 5 micromol/L, inhibited in both cultured PCa cells and DU145 xenografts (a) the expression of protein kinase Cepsilon (PKCepsilon), phosphatidylinositol 3-kinase, phosphorylated AKT, phosphorylated Janus-activated kinase-2, and phosphorylated signal transducer and activator of transcription 3 (Stat3); (b) the DNA-binding activity of transcription factors activator protein-1, nuclear factor-kappaB, and Stat3; and (c) Bcl-xL, cdc25A, and cyclooxygenase-2 expression. The results indicate for the first time, using both in vitro and in vivo preclinical models, that PL inhibits the growth and invasion of PCa. PL inhibits multiple molecular targets including PKCepsilon, a predictive biomarker of PCa aggressiveness. PL may be a novel agent for therapy of hormone-refractory PCa.


Subject(s)
Cell Division/drug effects , Naphthoquinones/pharmacology , Plants, Medicinal/chemistry , Prostatic Neoplasms/pathology , Animals , Apoptosis/drug effects , Base Sequence , DNA Primers , Electrophoretic Mobility Shift Assay , Humans , Immunohistochemistry , Male , Mice , Mice, Nude , Neoplasm Invasiveness
5.
Cancer Res ; 67(18): 8828-38, 2007 Sep 15.
Article in English | MEDLINE | ID: mdl-17875724

ABSTRACT

Prostate cancer is the most common type of cancer in men and ranks second only to lung cancer in cancer-related deaths. The management of locally advanced prostate cancer is difficult because the cancer often becomes hormone insensitive and unresponsive to current chemotherapeutic agents. Knowledge about the regulatory molecules involved in the transformation to androgen-independent prostate cancer is essential for the rational design of agents to prevent and treat prostate cancer. Protein kinase Cepsilon (PKCepsilon), a member of the novel PKC subfamily, is linked to the development of androgen-independent prostate cancer. PKCepsilon expression levels, as determined by immunohistochemistry of human prostate cancer tissue microarrays, correlated with the aggressiveness of prostate cancer. The mechanism by which PKCepsilon mediates progression to prostate cancer remains elusive. We present here for the first time that signal transducers and activators of transcription 3 (Stat3), which is constitutively activated in a wide variety of human cancers, including prostate cancer, interacts with PKCepsilon. The interaction of PKCepsilon with Stat3 was observed in human prostate cancer, human prostate cancer cell lines (LNCaP, DU145, PC3, and CW22rv1), and prostate cancer that developed in transgenic adenocarcinoma of mouse prostate mice. In reciprocal immunoprecipitation/blotting experiments, prostatic Stat3 coimmunoprecipitated with PKCepsilon. Localization of PKCepsilon with Stat3 was confirmed by double immunofluorescence staining. The interaction of PKCepsilon with Stat3 was PKCepsilon isoform specific. Inhibition of PKCepsilon protein expression in DU145 cells using specific PKCepsilon small interfering RNA (a) inhibited Stat3Ser727 phosphorylation, (b) decreased both Stat3 DNA-binding and transcriptional activity, and (c) decreased DU145 cell invasion. These results indicate that PKCepsilon activation is essential for constitutive activation of Stat3 and prostate cancer progression.


Subject(s)
Prostatic Neoplasms/metabolism , Protein Kinase C-epsilon/metabolism , STAT3 Transcription Factor/metabolism , Animals , Cell Line, Tumor , Cytokines/biosynthesis , Cytokines/genetics , Humans , Male , Mice , Mice, Inbred C57BL , Neoplasm Invasiveness , Phosphorylation , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Protein Kinase C-epsilon/biosynthesis , Protein Kinase C-epsilon/genetics , STAT3 Transcription Factor/biosynthesis , STAT3 Transcription Factor/genetics , Serine/metabolism , Signal Transduction , Transcription, Genetic , Transfection
6.
Cancer Res ; 64(21): 7756-65, 2004 Nov 01.
Article in English | MEDLINE | ID: mdl-15520180

ABSTRACT

Chronic exposure to UV radiation (UVR), especially in the UVA (315-400 nm) and UVB (280-315 nm) spectrum of sunlight, is the major risk factor for the development of nonmelanoma skin cancer. UVR is a complete carcinogen, which both initiates and promotes carcinogenesis. We found that protein kinase C epsilon (PKCepsilon), a member of the phospholipid-dependent threonine/serine kinase family, is an endogenous photosensitizer, the overexpression of which in the epidermis increases the susceptibility of mice to UVR-induced cutaneous damage and development of squamous cell carcinoma. The PKCepsilon transgenic mouse (FVB/N) lines 224 and 215 overexpressed 8- and 18-fold PKCepsilon protein, respectively, over endogenous levels in basal epidermal cells. UVR exposure (1 kJ/m(2) three times weekly) induced irreparable skin damage in high PKCepsilon-overexpressing mouse line 215. However, the PKCepsilon transgenic mouse line 224, when exposed to UVR (2 kJ/m(2) three times weekly), exhibited minimum cutaneous damage but increased squamous cell carcinoma multiplicity by 3-fold and decreased tumor latency by 12 weeks. UVR exposure of PKCepsilon transgenic mice compared with wild-type littermates (1) elevated the levels of neither cyclobutane pyrimidine dimer nor pyrimidine (6-4) pyrimidone dimer, (2) reduced the appearance of sunburn cells, (3) induced extensive hyperplasia and increased the levels of mouse skin tumor promoter marker ornithine decarboxylase, and (4) elevated the levels of tumor necrosis factor alpha (TNFalpha) and other growth stimulatory cytokines, granulocyte colony-stimulating factor, and granulocyte macrophage colony-stimulating factor. The role of TNFalpha in UVR-induced cutaneous damage was evaluated using PKCepsilon transgenic mice deficient in TNFalpha. UVR treatment three times weekly for 13 weeks at 2 kJ/m(2) induced severe cutaneous damage in PKCepsilon transgenic mice (line 215), which was partially prevented in PKCepsilon-transgenic TNFalpha-knockout mice. Taken together, the results indicate that PKCepsilon signals UVR-induced TNFalpha release that is linked, at least in part, to the photosensitivity of PKCepsilon transgenic mice.


Subject(s)
Carcinoma, Squamous Cell/etiology , Neoplasms, Radiation-Induced/etiology , Protein Kinase C/physiology , Skin Neoplasms/etiology , Skin/radiation effects , Ultraviolet Rays , Animals , Cytokines/biosynthesis , Mice , Mice, Transgenic , Ornithine Decarboxylase/genetics , Proliferating Cell Nuclear Antigen/analysis , Protein Kinase C-epsilon , Radiation Tolerance , Tumor Necrosis Factor-alpha/physiology
7.
Oncogene ; 21(22): 3620-30, 2002 May 16.
Article in English | MEDLINE | ID: mdl-12032864

ABSTRACT

Protein Kinase Cdelta (PKCdelta), a Ca(2+)-independent, phospholipid-dependent serine/threonine kinase, is among the PKC isoforms expressed in mouse epidermis. We reported that FVB/N transgenic mice that overexpress ( approximately eightfold) PKCdelta protein in basal epidermal cells are resistant to skin tumor formation by the 7,12-dimethylbenz(a)anthracene (DMBA)-initiation and 12-O-tetradecanoylphorbol-13-acetate (TPA)-promotion protocol. However, despite being resistant to skin tumor promotion by TPA, PKCdelta transgenic mice elicited a 3-4-fold increase in TPA-induced epidermal ODC activity and putrescine levels than their wild-type littermates. PKCdelta was observed to be the key component of the TPA signal transduction pathways to the induction of mouse epidermal ODC activity. To determine if TPA-induced ODC activity and associated putrescine levels in PKCdelta transgenic mice contributed to PKCdelta-mediated suppression of skin tumor promotion by TPA, the irreversible inhibitor of ODC, alpha-difluoromethylornithine (DFMO), was used. PKCdelta transgenic mice and their wild-type littermates were initiated with 100 nmol DMBA and then promoted twice weekly with 5 nmol TPA. The experimental group was given 0.5% DFMO in their drinking water, while the control group was given tap water. After 25 weeks, the number of papillomas (>2 mm) per mouse was counted. The DFMO treatment did not affect the skin tumor multiplicity of PKCdelta transgenic mice. These results indicate that PKCdelta-induced ODC activity is not involved in PKCdelta-mediated tumor suppression. Thus, the signaling pathways via PKCdelta to epidermal ODC induction and skin tumor suppression appear to be independent.


Subject(s)
Isoenzymes/physiology , Ornithine Decarboxylase/metabolism , Protein Kinase C/physiology , Skin Neoplasms/enzymology , 9,10-Dimethyl-1,2-benzanthracene/antagonists & inhibitors , Animals , Carcinogens/antagonists & inhibitors , Carcinogens/pharmacology , Cell Cycle Proteins/metabolism , Cell Division , Eflornithine/pharmacology , Enzyme Activation , Enzyme Induction , Enzyme Inhibitors/pharmacology , Epidermis/enzymology , Epidermis/pathology , Female , Isoenzymes/genetics , Kinetics , Male , Mice , Mice, Knockout , Mice, Transgenic , Ornithine Decarboxylase/genetics , Ornithine Decarboxylase Inhibitors , Protein Kinase C/genetics , Protein Kinase C-delta , RNA, Messenger/biosynthesis , Signal Transduction , Skin Neoplasms/chemically induced , Skin Neoplasms/pathology , Tetradecanoylphorbol Acetate/antagonists & inhibitors , Tetradecanoylphorbol Acetate/pharmacology , Tumor Suppressor Proteins/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...