101
|
Tabe Y, Konopleva M, Andreeff M, Ohsaka A. Effects of PPARγ Ligands on Leukemia. PPAR Res 2012; 2012:483656. [PMID: 22685453 PMCID: PMC3364693 DOI: 10.1155/2012/483656] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2012] [Accepted: 03/21/2012] [Indexed: 12/18/2022] Open
Abstract
Peroxisome proliferator-activated receptors (PPARs) and retinoic acid receptors (RARs), members of the nuclear receptor superfamily, are transcription factors that regulate a variety of important cellular functions. PPARs form heterodimers retinoid X receptor (RXR), an obligate heterodimeric partner for other nuclear receptors. Several novel links between retinoid metabolism and PPAR responses have been identified, and activation of PPAR/RXR expression has been shown to increase response to retinoids. PPARγ has emerged as a key regulator of cell growth and survival, whose activity is modulated by a number of synthetic and natural ligands. While clinical trials in cancer patients with thiazolidinediones (TZD) have been disappointing, novel structurally different PPARγ ligands, including triterpenoids, have entered clinical arena as therapeutic agents for epithelial and hematopoietic malignancies. Here we shall review the antitumor advances of PPARγ, alone and in combination with RARα ligands in control of cell proliferation, differentiation, and apoptosis and their potential therapeutic applications in hematological malignancies.
Collapse
Affiliation(s)
- Yoko Tabe
- Department of Clinical Laboratory Medicine, Juntendo University School of Medicine, Hongo 2-1-1, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Marina Konopleva
- Department of Leukemia, The University of Texas M.D. Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Michael Andreeff
- Department of Leukemia, The University of Texas M.D. Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Akimichi Ohsaka
- Department of Transfusion Medicine and Stem Cell Regulation, Juntendo University School of Medicine, Hongo 2-1-1, Bunkyo-ku, Tokyo 113-8421, Japan
| |
Collapse
|
102
|
Vamecq J, Colet JM, Vanden Eynde JJ, Briand G, Porchet N, Rocchi S. PPARs: Interference with Warburg' Effect and Clinical Anticancer Trials. PPAR Res 2012; 2012:304760. [PMID: 22654896 PMCID: PMC3357561 DOI: 10.1155/2012/304760] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2011] [Revised: 02/15/2012] [Accepted: 02/19/2012] [Indexed: 02/07/2023] Open
Abstract
The metabolic/cell signaling basis of Warburg's effect ("aerobic glycolysis") and the general metabolic phenotype adopted by cancer cells are first reviewed. Several bypasses are adopted to provide a panoramic integrated view of tumoral metabolism, by attributing a central signaling role to hypoxia-induced factor (HIF-1) in the expression of aerobic glycolysis. The cancer metabolic phenotype also results from alterations of other routes involving ras, myc, p53, and Akt signaling and the propensity of cancer cells to develop signaling aberrances (notably aberrant surface receptor expression) which, when present, offer unique opportunities for therapeutic interventions. The rationale for various emerging strategies for cancer treatment is presented along with mechanisms by which PPAR ligands might interfere directly with tumoral metabolism and promote anticancer activity. Clinical trials using PPAR ligands are reviewed and followed by concluding remarks and perspectives for future studies. A therapeutic need to associate PPAR ligands with other anticancer agents is perhaps an important lesson to be learned from the results of the clinical trials conducted to date.
Collapse
Affiliation(s)
- Joseph Vamecq
- Inserm, HMNO, CBP, CHRU Lille, 59037 Lille, France
- Biochemistry and Molecular Biology, HMNO, CBP, CHRU Lille, 59037 Lille, France
| | - Jean-Marie Colet
- Department of Human Biology and Toxicology, Faculty of Medicine and Pharmacy, UMons, 7000 Mons, Belgium
| | | | - Gilbert Briand
- Biochemistry and Molecular Biology, HMNO, CBP, CHRU Lille, 59037 Lille, France
| | - Nicole Porchet
- Biochemistry and Molecular Biology, HMNO, CBP, CHRU Lille, 59037 Lille, France
| | - Stéphane Rocchi
- Inserm U1065, IFR 50, Mediterranean Center of Molecular Medicine, 06204 Nice, France
| |
Collapse
|
103
|
Tanaka T, Shnimizu M, Moriwaki H. Cancer chemoprevention by carotenoids. Molecules 2012; 17:3202-42. [PMID: 22418926 PMCID: PMC6268471 DOI: 10.3390/molecules17033202] [Citation(s) in RCA: 317] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2011] [Revised: 02/15/2012] [Accepted: 03/06/2012] [Indexed: 02/07/2023] Open
Abstract
Carotenoids are natural fat-soluble pigments that provide bright coloration to plants and animals. Dietary intake of carotenoids is inversely associated with the risk of a variety of cancers in different tissues. Preclinical studies have shown that some carotenoids have potent antitumor effects both in vitro and in vivo, suggesting potential preventive and/or therapeutic roles for the compounds. Since chemoprevention is one of the most important strategies in the control of cancer development, molecular mechanism-based cancer chemoprevention using carotenoids seems to be an attractive approach. Various carotenoids, such as β-carotene, a-carotene, lycopene, lutein, zeaxanthin, β-cryptoxanthin, fucoxanthin, canthaxanthin and astaxanthin, have been proven to have anti-carcinogenic activity in several tissues, although high doses of β-carotene failed to exhibit chemopreventive activity in clinical trials. In this review, cancer prevention using carotenoids are reviewed and the possible mechanisms of action are described.
Collapse
Affiliation(s)
- Takuji Tanaka
- Tohkai Cytopathology Institute, Cancer Research and Prevention-TCI-CaRP, 5-1-2 Minami-Uzura, Gifu 500-8285, Japan.
| | | | | |
Collapse
|
104
|
The PPAR Gamma Agonist Troglitazone Regulates Erk 1/2 Phosphorylation via a PPARγ-Independent, MEK-Dependent Pathway in Human Prostate Cancer Cells. PPAR Res 2012; 2012:929052. [PMID: 22448169 PMCID: PMC3289875 DOI: 10.1155/2012/929052] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2011] [Revised: 11/22/2011] [Accepted: 11/23/2011] [Indexed: 12/20/2022] Open
Abstract
Thiazolidinediones (TZDs) dramatically reduce the growth of human prostate cancer cells in vitro and in vivo. To determine whether the antitumor effects of TZDs were due in part to changes in the MEK/Erk signaling pathway, we examined the regulation of Erk phosphorylation by the TZD troglitazone within the PC-3 and C4-2 human prostate cancer cell lines. Western blot analysis revealed troglitazone-induced phosphorylation of Erk in both PC-3 and C4-2 cells. Troglitazone-induced increases in Erk phosphorylation were suppressed by the MEK inhibitor U0126 but not by the PPARγ antagonist GW9662. Pretreatment with U0126 did not alter the ability of troglitazone to regulate expression of two proteins that control cell cycle, p21, and c-Myc. Troglitazone was also still effective at reducing PC-3 proliferation in the presence of U0126. Therefore, our data suggest that troglitazone-induced Erk phosphorylation does not significantly contribute to the antiproliferative effect of troglitazone.
Collapse
|
105
|
Peters JM, Shah YM, Gonzalez FJ. The role of peroxisome proliferator-activated receptors in carcinogenesis and chemoprevention. Nat Rev Cancer 2012; 12:181-95. [PMID: 22318237 PMCID: PMC3322353 DOI: 10.1038/nrc3214] [Citation(s) in RCA: 375] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Peroxisome proliferator-activated receptors (PPARs) are ligand-activated transcription factors that are involved in regulating glucose and lipid homeostasis, inflammation, proliferation and differentiation. Although all of these functions might contribute to the influence of PPARs in carcinogenesis, there is a distinct need for a review of the literature and additional experimentation to determine the potential for targeting PPARs for cancer therapy and cancer chemoprevention. As PPAR agonists include drugs that are used for the treatment of metabolic diseases, a more complete understanding of the roles of PPARs in cancer will aid in determining any increased cancer risk for patients undergoing therapy with PPAR agonists.
Collapse
Affiliation(s)
- Jeffrey M Peters
- Department of Veterinary and Biomedical Sciences and The Center for Molecular Toxicology and Carcinogenesis, The Pennsylvania State University, University Park, Pennsylvania 16802, USA.
| | | | | |
Collapse
|
106
|
Abstract
In this article we review the evolution of cancer research involving PPARgamma, including mechanisms, target genes, and clinical applications. For the last thirteen years, the effects of PPARgamma activity on tumor biology have been studied intensely. Most of this research has focused upon the potential for employing agonists of this nuclear receptor in cancer treatment. As a monotherapy such agonists have shown little success in clinical trials, while they have shown promise as components of combination treatments both in culture and in animal models. Other investigations have explored a possible role for PPARgamma as a tumor suppressor, and as an inducer of differentiation of cancer stem cells. Whereas early studies have yielded variable conclusions regarding the prevalence of PPARgamma mutations in cancer, the protein level of this receptor has been more recently identified as a significant prognostic marker. We predict that indicators of PPARgamma activity may also serve as predictive markers for tailoring treatments.
Collapse
Affiliation(s)
- Gregory T Robbins
- Department of Medical Microbiology, Immunology and Cell Biology, Southern Illinois University School of Medicine
| | | |
Collapse
|
107
|
Abdelbaqi K, Lack N, Guns ET, Kotha L, Safe S, Sanderson JT. Antiandrogenic and growth inhibitory effects of ring-substituted analogs of 3,3'-diindolylmethane (ring-DIMs) in hormone-responsive LNCaP human prostate cancer cells. Prostate 2011; 71:1401-1412. [PMID: 21321979 DOI: 10.1002/pros.21356] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2010] [Accepted: 01/14/2011] [Indexed: 11/09/2022]
Abstract
BACKGROUND Cruciferous vegetables protect against prostate cancer. Indole-3-carbinol (I3C) and its major metabolite 3,3'-diindolylmethane (DIM), exhibit antitumor activities in vitro and in vivo. Several synthetic ring-substituted dihaloDIMs (ring-DIMs) appear to have increased anticancer activity. METHODS Inhibition of LNCaP prostate cancer cell growth was measured by a WST-1 cell viability assay. Cytoplasmic and nuclear proteins were analyzed by immunoblotting and immunofluorescence. Androgen receptor (AR) activation was assessed by measuring prostate-specific antigen (PSA) expression and using LNCaP cells containing human AR and an AR-dependent probasin promoter-green fluorescent protein (GFP) construct. RESULTS Like DIM, several ring-substituted dihaloDIM analogs, namely 4,4'-dibromo-, 4,4'-dichloro-, 7,7'-dibromo-, and 7,7'-dichloroDIM, significantly inhibited DHT-stimulated growth of LNCaP cells at concentrations ≥1 µM. We observed structure-dependent differences for the effects of the ring-DIMs on AR expression, nuclear AR accumulation and PSA levels in LNCaP cells after 24 hr. Both 4,4'- and 7,7'-dibromoDIM decreased AR protein and mRNA levels, whereas 4,4'- and 7,7'-dichloroDIM had minimal effect. All four dihaloDIMs (10 and 30 µM) significantly decreased PSA protein and mRNA levels. Immuofluorescence studies showed that only the dibromoDIMs increased nuclear localization of AR. All ring-DIMs caused a concentration-dependent decrease in fluorescence induced by the synthetic androgen R1881 in LNCaP cells transfected with wild-type human AR and an androgen-responsive probasin promoter-GFP gene construct, with potencies up to 10-fold greater than that of DIM. CONCLUSION The antiandrogenic effects of ring-DIMs suggest they may form the basis for the development of novel agents against hormone-sensitive prostate cancer, alone or in combination with other drugs.
Collapse
Affiliation(s)
- Khalil Abdelbaqi
- INRS-Institut Armand-Frappier, Université du Québec, Laval, QC, Canada
| | | | | | | | | | | |
Collapse
|
108
|
Venkatachalam G, Kumar AP, Sakharkar KR, Thangavel S, Clement MV, Sakharkar MK. PPARγ disease gene network and identification of therapeutic targets for prostate cancer. J Drug Target 2011; 19:781-96. [PMID: 21780947 DOI: 10.3109/1061186x.2011.568062] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|
109
|
CXCR4 in Cancer and Its Regulation by PPARgamma. PPAR Res 2011; 2008:769413. [PMID: 18779872 PMCID: PMC2528256 DOI: 10.1155/2008/769413] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2008] [Revised: 06/25/2008] [Accepted: 07/10/2008] [Indexed: 12/20/2022] Open
Abstract
Chemokines are peptide mediators involved in normal development,
hematopoietic and immune regulation, wound healing, and
inflammation. Among the chemokines is CXCL12, which binds
principally to its receptor CXCR4 and regulates leukocyte
precursor homing to bone marrow and other sites. This role of
CXCL12/CXCR4 is “commandeered” by cancer cells to facilitate the
spread of CXCR4-bearing tumor cells to tissues with high CXCL12
concentrations. High CXCR4 expression by cancer cells predisposes
to aggressive spread and metastasis and ultimately to poor patient
outcomes. As well as being useful as a marker for disease
progression, CXCR4 is a potential target for anticancer therapies.
It is possible to interfere directly with the CXCL12:CXCR4 axis
using peptide or small-molecular-weight antagonists. A further
opportunity is offered by promoting strategies that downregulate
CXCR4 pathways: CXCR4 expression in the tumor microenvironment is
modulated by factors such as hypoxia, nucleosides, and
eicosanoids. Another promising approach is through targeting PPAR
to suppress CXCR4 expression. Endogenous PPARγ such as 15-deoxy-Δ12,14-PGJ2 and synthetic agonists such as the
thiazolidinediones both cause downregulation of CXCR4 mRNA and
receptor. Adjuvant therapy using PPARγ agonists may, by
stimulating PPARγ-dependent downregulation of CXCR4 on cancer cells, slow the rate of metastasis and impact beneficially on
disease progression.
Collapse
|
110
|
PPARgamma: The Portrait of a Target Ally to Cancer Chemopreventive Agents. PPAR Res 2011; 2008:436489. [PMID: 18779870 PMCID: PMC2528242 DOI: 10.1155/2008/436489] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2008] [Revised: 05/22/2008] [Accepted: 07/16/2008] [Indexed: 12/13/2022] Open
Abstract
Peroxisome proliferator-activated receptor-gamma (PPARγ), one of three ligand-activated transcription factors named PPAR, has been identified as a molecular target for cancer chemopreventive agents. PPARγ was initially understood as a regulator of adipocyte differentiation and glucose homeostasis while later on, it became evident that it is also involved in cell differentiation, apoptosis, and angiogenesis, biological processes which are deregulated in cancer. It is now established that PPARγ ligands can induce cell differentiation and yield early antineoplastic effects in several tumor types. Moreover, several bioactive natural products with cancer protecting potential are shown to operate through activation of PPARγ. Overall, PPARγ appears to be a prevalent target ally to cancer chemopreventive agents and therefore pursuing research in this area is of great relevance.
Collapse
|
111
|
To Live or to Die: Prosurvival Activity of PPARgamma in Cancers. PPAR Res 2011; 2008:209629. [PMID: 18784849 PMCID: PMC2532487 DOI: 10.1155/2008/209629] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2008] [Accepted: 05/03/2008] [Indexed: 11/21/2022] Open
Abstract
The role of PPARγ in tumorigenesis is controversial. In this article, we review and analyze literature from the past decade that highlights the potential proneoplastic activity of PPARγ. We discuss the following five aspects of the nuclear hormone receptor and its agonists: (1) relative expression of PPARγ in human tumor versus normal tissues; (2) receptor-dependent proneoplastic effects; (3) impact of PPARγ and its agonists on tumors in animal models; (4) clinical trials of thiazolidinediones (TZDs) in human malignancies; (5) TZDs as chemopreventive agents in epidemiology studies. The focus is placed on the most relevant in vivo animal models and human data. In vitro cell line studies are included only when the effects are shown to be dependent on the PPARγ receptor.
Collapse
|
112
|
Abstract
Peroxisome proliferators-activated receptors (PPARs) that are members of the nuclear receptor superfamily have three different isoforms: PPARalpha, PPARdelta, and PPARgamma. PPARs are ligand-activated transcription factors, and they are implicated in tumor progression, differentiation, and apoptosis. Activation of PPAR isoforms lead to both anticarcinogenesis and anti-inflammatory effect. It has so far identified many PPAR ligands including chemical composition and natural occurring. PPAR ligands are reported to activate PPAR signaling and exert cancer prevention and treatment in vitro and/or in vivo studies. Although the effects depend on the isoforms and the types of ligands, biological modulatory activities of PPARs in carcinogenesis and disease progression are attracted for control or combat cancer development. This short review summarizes currently available data on the role of PPAR ligands in carcinogenesis.
Collapse
|
113
|
Potential of peroxisome proliferator-activated receptor gamma antagonist compounds as therapeutic agents for a wide range of cancer types. PPAR Res 2011; 2008:494161. [PMID: 18779871 PMCID: PMC2528255 DOI: 10.1155/2008/494161] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2008] [Accepted: 06/09/2008] [Indexed: 01/31/2023] Open
Abstract
PPARγ is a therapeutic target that has been exploited for
treatment of type II diabetes mellitus (T2DM) with agonist drugs.
Since PPARγ is expressed by many hematopoietic, mesodermal and
epithelial cancers, agonist drugs were tested and shown to have
both preclinical and clinical anticancer activities. While
preclinical activity has been observed in many cancer types,
clinical activity has been observed only in pilot and phase II
trials in liposarcoma and prostate cancer. Most studies address
agonist compounds, with substantially fewer reports on anticancer
effects of PPARγ antagonists. In cancer model systems, some
effects of PPARγ agonists were not inhibited by PPARγ antagonists,
suggesting noncanonical or PPARγ-independent mechanisms. In
addition, PPARγ antagonists, such as T0070907 and GW9662, have
exhibited antiproliferative effects on a broad range of
hematopoietic and epithelial cell lines, usually with greater
potency than agonists. Also, additive antiproliferative effects
of combinations of agonist plus antagonist drugs were observed.
Finally, there are preclinical in vivo data showing that
antagonist compounds can be administered safely, with favorable
metabolic effects as well as antitumor effects. Since PPARγ
antagonists represent a new drug class that holds promise as a
broadly applicable therapeutic approach for cancer treatment, it
is the subject of this review.
Collapse
|
114
|
Multiple Interactions between Peroxisome Proliferators-Activated Receptors and the Ubiquitin-Proteasome System and Implications for Cancer Pathogenesis. PPAR Res 2011; 2008:195065. [PMID: 18551186 PMCID: PMC2423003 DOI: 10.1155/2008/195065] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2008] [Accepted: 04/29/2008] [Indexed: 12/31/2022] Open
Abstract
The peroxisome proliferator-activated receptors (PPAR) α, β/δ, and γ are ligand-activated nuclear receptors involved in a number of physiological processes, including lipid and glucose homeostasis, inflammation, cell growth, differentiation, and death. PPAR agonists are used in the treatment of human diseases, like type 2 diabetes and dyslipidemia, and PPARs appear as promising therapeutic targets in other conditions, including cancer. A better understanding of the functions and regulation of PPARs in normal and pathological processes is of primary importance to devise appropriate therapeutic strategies. The ubiquitin-proteasome system (UPS) plays an important role in controlling level and activity of many nuclear receptors and transcription factors. PPARs are subjected to UPS-dependent regulation. Interestingly, the three PPAR isotypes are differentially regulated by the UPS in response to ligand-dependent activation, a phenomenon that may be intrinsically connected to their distinct cellular functions and behaviors. In addition to their effects ongene expression, PPARs appear to affect protein levels and downstream pathways also by modulating the activity of the UPS in target-specific manners. Here we review the current knowledge of the interactions between the UPS and PPARs in light of the potential implications for their effects on cell fate and tumorigenesis.
Collapse
|
115
|
Role of peroxisome proliferator-activated receptor gamma and its ligands in the treatment of hematological malignancies. PPAR Res 2011; 2008:834612. [PMID: 18528522 PMCID: PMC2408681 DOI: 10.1155/2008/834612] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2008] [Accepted: 04/21/2008] [Indexed: 02/07/2023] Open
Abstract
Peroxisome proliferator-activated receptor gamma (PPARgamma) is a multifunctional transcription factor with important regulatory roles in inflammation, cellular growth, differentiation, and apoptosis. PPARgamma is expressed in a variety of immune cells as well as in numerous leukemias and lymphomas. Here, we review recent studies that provide new insights into the mechanisms by which PPARgamma ligands influence hematological malignant cell growth, differentiation, and survival. Understanding the diverse properties of PPARgamma ligands is crucial for the development of new therapeutic approaches for hematological malignancies.
Collapse
|
116
|
Chondrosarcoma and peroxisome proliferator-activated receptor. PPAR Res 2011; 2008:250568. [PMID: 18725985 PMCID: PMC2517661 DOI: 10.1155/2008/250568] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2008] [Accepted: 07/17/2008] [Indexed: 12/20/2022] Open
Abstract
Induction of differentiation and apoptosis in cancer cells by ligands of PPARγ is a novel therapeutic approach to malignant tumors. Chondrosarcoma (malignant cartilage tumor) and OUMS-27 cells (cell line established from grade III human chondrosarcoma) express PPARγ. PPARγ ligands inhibited cell proliferation in a dose-dependent manner, and induced apoptosis of OUMS-27. The higher-grade chondrosarcoma expressed a higher amount of antiapoptotic Bcl-xL in vivo. The treatment of OUMS-27 by 15d-PGJ2, the most potent endogenous ligand for PPARγ, downregulated expression of Bcl-xL and induced transient upregulation of proapoptotic Bax, which could accelerate cytochrome c release from mitochondria to the cytosol, followed by induction of caspase-dependent apoptosis. 15d-PGJ2 induced the expression of CDK inhibitor p21 protein in human chondrosarcoma cells, which appears to be involved in the mechanism of inhibition of cell proliferation. These findings suggest that targeted therapy with PPARγ ligands could be a novel strategy against chondrosarcoma.
Collapse
|
117
|
Omega-3 Fatty Acids and PPARgamma in Cancer. PPAR Res 2011; 2008:358052. [PMID: 18769551 PMCID: PMC2526161 DOI: 10.1155/2008/358052] [Citation(s) in RCA: 91] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2008] [Revised: 05/30/2008] [Accepted: 06/24/2008] [Indexed: 01/25/2023] Open
Abstract
Omega-3 (or n-3) polyunsaturated fatty acids (PUFAs) and their metabolites are natural ligands for peroxisome proliferator receptor activator (PPAR)gamma and, due to the effects of PPARgamma on cell proliferation, survival, and differentiation, are potential anticancer agents. Dietary intake of omega-3 PUFAs has been associated with a reduced risk of certain cancers in human populations and in animal models. In vitro studies have shown that omega-3 PUFAs inhibit cell proliferation and induce apoptosis in cancer cells through various pathways but one of which involves PPARgamma activation. The differential activation of PPARgamma and PPARgamma-regulated genes by specific dietary fatty acids may be central to their distinct roles in cancer. This review summarizes studies relating PUFAs to PPARgamma and cancer and offers a new paradigm relating an n-3 PUFA through PPARgamma to the expression of the cell surface proteoglycan, syndecan-1, and to the death of cancer cells.
Collapse
|
118
|
Abstract
Peroxisome proliferator-activated receptors (PPARs) are ligand-activated transcription factors that belong to the nuclear hormone receptor superfamily. PPARalpha is mainly expressed in the liver, where it activates fatty acid catabolism. PPARalpha activators have been used to treat dyslipidemia, causing a reduction in plasma triglyceride and elevation of high-density lipoprotein cholesterol. PPARdelta is expressed ubiquitously and is implicated in fatty acid oxidation and keratinocyte differentiation. PPARdelta activators have been proposed for the treatment of metabolic disease. PPARgamma2 is expressed exclusively in adipose tissue and plays a pivotal role in adipocyte differentiation. PPARgamma is involved in glucose metabolism through the improvement of insulin sensitivity and represents a potential therapeutic target of type 2 diabetes. Thus PPARs are molecular targets for the development of drugs treating metabolic syndrome. However, PPARs also play a role in the regulation of cancer cell growth. Here, we review the function of PPARs in tumor growth.
Collapse
|
119
|
Li H, Li QD, Wang PZ, Wang MS, Cui J, Diao TY, Li QH. The effect of oxidized low-density lipoprotein combined with adriamycin on the proliferation of Eca-109 cell line. Lipids Health Dis 2011; 10:108. [PMID: 21711568 PMCID: PMC3150309 DOI: 10.1186/1476-511x-10-108] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2011] [Accepted: 06/29/2011] [Indexed: 01/19/2023] Open
Abstract
Background The purpose of this study was to identify the affect on the proliferation Eca-109 cells treated with oxidized low-density lipoprotein (ox-LDL) combined with adriamycin (ADM). Methods Eca-109 cell were cultured in the presence of oxLDL/ADM, and cell proliferation tested by MTT and cell apoptosis was monitored by the proportion of apoptosis and cell cycle by flow cytomester. We simultaneously evaluated the level of associated- apoptosis Bcl-2, Bax, and Caspase-3 gene mRNA and protein. Results OxLDL were cytotoxic and activate apoptosis. OxLDL combined with ADM significant enhanced the proportion rate of apoptosis on a time and dose dependency. The expressions of the inhibiting apoptosis Bcl-2 gene mRNA and protein were down regulated, whereas, the expressions of the promoting apoptosis Bax, and Caspase-3 genes mRNA and protein were up regulation. Conclusion These results suggested that oxLDL have cytotoxicity and activate apoptosis on the Eca-109 cells. OxLDL combined with ADM have a synergistic effect on the apoptosis induced Eca-109 cells. Furthermore, oxLDL may contribute to the improvement of clinical chemotherapy of cancer need to make further investigation.
Collapse
Affiliation(s)
- Hao Li
- Qilu Hospital, Shandong University, Jinan, P.R. China.
| | | | | | | | | | | | | |
Collapse
|
120
|
Akinyeke TO, Stewart LV. Troglitazone suppresses c-Myc levels in human prostate cancer cells via a PPARγ-independent mechanism. Cancer Biol Ther 2011; 11:1046-58. [PMID: 21525782 DOI: 10.4161/cbt.11.12.15709] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Troglitazone is a ligand for the peroxisome proliferator activated receptor gamma (PPARγ) that decreases growth of human prostate cancer cells in vitro and in vivo. However, the mechanism by which troglitazone reduces prostate cancer cell growth is not fully understood. To understand the signaling pathways involved in troglitazone-induced decreases in prostate cancer growth, we examined the effect of troglitazone on androgen-independent C4-2 human prostate cancer cells. Initial experiments revealed troglitazone inhibited C4-2 cell proliferation by arresting cells in the G(0)/G(1) phase of the cell cycle and inducing apoptosis. Since the proto-oncogene product c-Myc regulates both apoptosis and cell cycle progression, we next examined whether troglitazone altered expression of c-Myc. Troglitazone decreased c-Myc protein levels as well as expression of downstream targets of c-Myc in a dose-dependent manner. In C4-2 cells, troglitazone-induced decreases in c-Myc protein involve proteasome-mediated degradation of c-Myc protein as well as reductions in c-Myc mRNA levels. It appears that troglitazone stimulates degradation of c-Myc by increasing c-Myc phosphorylation, for the level of phosphorylated c-Myc was elevated in prostate cancer cells exposed to troglitazone. While troglitazone dramatically decreased the amount of c-Myc within C4-2 cells, the PPARγ ligands ciglitazone, rosiglitazone and pioglitazone did not reduce c-Myc protein levels. Furthermore the down-regulation of c-Myc by troglitazone was not blocked by the PPARγ antagonist GW9662 and siRNA-mediated decreases in PPARγ protein. Thus, our data suggest that troglitazone reduces c-Myc protein independently of PPARγ.
Collapse
Affiliation(s)
- Tunde O Akinyeke
- Department of Biochemistry and Cancer Biology, Meharry Medical College, Nashville, TN, USA
| | | |
Collapse
|
121
|
Cannata D, Fierz Y, Vijayakumar A, LeRoith D. Type 2 diabetes and cancer: what is the connection? ACTA ACUST UNITED AC 2011; 77:197-213. [PMID: 20309918 DOI: 10.1002/msj.20167] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Epidemiological studies have demonstrated an association between type 2 diabetes and cancer. Type 2 diabetes is characterized by insulin resistance and hyperinsulinemia. Hyperinsulinemia may lead to cancer through insulin's effect on its cognate receptor and the insulin-like growth factor system. The effects of insulin and insulin-like growth factor I on cancer development and progression have been demonstrated in animal and human studies. Type 2 diabetes has been positively associated with cancers of the breast, colon, and pancreas. An inverse relationship has been observed between type 2 diabetes and prostate cancer, and this may be due to lower testosterone levels in men with type 2 diabetes. Medications used to treat type 2 diabetes may affect cancer cells directly or indirectly by affecting serum insulin levels. Hyperinsulinemia may be an important risk factor for cancer as well as a target for cancer therapy.
Collapse
Affiliation(s)
- Dara Cannata
- Mount Sinai School of Medicine, New York, NY, USA
| | | | | | | |
Collapse
|
122
|
Cerquetti L, Sampaoli C, Amendola D, Bucci B, Masuelli L, Marchese R, Misiti S, De Venanzi A, Poggi M, Toscano V, Stigliano A. Rosiglitazone induces autophagy in H295R and cell cycle deregulation in SW13 adrenocortical cancer cells. Exp Cell Res 2011; 317:1397-410. [DOI: 10.1016/j.yexcr.2011.02.014] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2010] [Revised: 02/17/2011] [Accepted: 02/25/2011] [Indexed: 10/18/2022]
|
123
|
Campbell SE, Rudder B, Phillips RB, Whaley SG, Stimmel JB, Leesnitzer LM, Lightner J, Dessus-Babus S, Duffourc M, Stone WL, Menter DG, Newman RA, Yang P, Aggarwal BB, Krishnan K. γ-Tocotrienol induces growth arrest through a novel pathway with TGFβ2 in prostate cancer. Free Radic Biol Med 2011; 50:1344-54. [PMID: 21335085 DOI: 10.1016/j.freeradbiomed.2011.02.007] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2010] [Revised: 02/07/2011] [Accepted: 02/09/2011] [Indexed: 12/24/2022]
Abstract
Regions along the Mediterranean and in southern Asia have lower prostate cancer incidence compared to the rest of the world. It has been hypothesized that one of the potential contributing factors for this low incidence includes a higher intake of tocotrienols. Here we examine the potential of γ-tocotrienol (GT3) to reduce prostate cancer proliferation and focus on elucidating pathways by which GT3 could exert a growth-inhibitory effect on prostate cancer cells. We find that the γ and δ isoforms of tocotrienol are more effective at inhibiting the growth of prostate cancer cell lines (PC-3 and LNCaP) compared with the γ and δ forms of tocopherol. Knockout of PPAR-γ and GT3 treatment show inhibition of prostate cancer cell growth, through a partially PPAR-γ-dependent mechanism. GT3 treatment increases the levels of the 15-lipoxygenase-2 enzyme, which is responsible for the conversion of arachidonic acid to the PPAR-γ-activating ligand 15-S-hydroxyeicosatrienoic acid. In addition, the latent precursor and the mature forms of TGFβ2 are down-regulated after treatment with GT3, with concomitant disruptions in TGFβ receptor I, SMAD-2, p38, and NF-κB signaling.
Collapse
Affiliation(s)
- Sharon E Campbell
- Department of Biochemistry and Molecular Biology, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
124
|
He XX, Tu SM, Lee MH, Yeung SCJ. Thiazolidinediones and metformin associated with improved survival of diabetic prostate cancer patients. Ann Oncol 2011; 22:2640-2645. [PMID: 21415239 DOI: 10.1093/annonc/mdr020] [Citation(s) in RCA: 127] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND The association between antidiabetic medications and the prognosis of human prostate cancer has not been explored. This study examined the impact of these drugs on the outcomes of diabetic patients with prostate cancer to provide a basis for diabetes management strategy in these patients. PATIENTS AND METHODS Records of consecutive prostate cancer patients with coexisting diabetes mellitus type 2 who were treated at the study institution between 15 July 1999 and 31 December 2008 were reviewed. The survival, cancer pathological grade, stage at the time of diagnosis, and antidiabetic pharmacotherapy of the patients were analyzed. RESULTS A total of 233 consecutive cases were analyzed. In Kaplan-Meier analysis, thiazolidinedione (log-rank, P = 0.005) and metformin (log-rank, P = 0.035) usage were significant predictors of improved overall survival, while insulin and insulin secretagogue usage were not significant predictors. Multivariate Cox regression analysis showed that thiazolidinedione {hazard ratio [HR] = 0.454 [95% confidence interval (CI) 0.213-0.965], P = 0.040} and metformin [HR = 0.550 (95% CI 0.315-0.960), P = 0.035] usage remained as significant predictors of favorable survival after controlling for variables including age, race, Gleason grade, and stage. CONCLUSIONS Thiazolidinediones and metformin appear to be associated with improved overall survival of diabetic prostate cancer patients. The choice of antidiabetic pharmacotherapy may influence overall survival of these patients.
Collapse
Affiliation(s)
- X-X He
- Department of Molecular and Cellular Oncology; Emergency Medicine
| | - S M Tu
- Genitourinary Medical Oncology
| | - M-H Lee
- Department of Molecular and Cellular Oncology
| | - S-C J Yeung
- Emergency Medicine; Endocrine Neoplasia and Hormonal Disorders, The University of Texas MD Anderson Cancer Center, Houston, USA.
| |
Collapse
|
125
|
Zhu H, Han B, Pan X, Qi H, Xu L. Thiazolidenediones induce tumour-cell apoptosis through the Akt-GSK3β pathway. J Clin Pharm Ther 2011; 37:65-70. [PMID: 21410737 DOI: 10.1111/j.1365-2710.2011.01251.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
WHAT IS KNOWN AND OBJECTIVE Prostate cancer is a major health threat for men. Thiazolidenediones (TZDs) are synthetic ligands of the peroxisome proliferator-activated receptor γ (PPARγ), and previous studies have shown that TZDs induce apoptosis of prostate cancer cells independently of PPARγ activation. However, the exact mechanism of these effects remains unknown. Our objective was to investigate the effects of TZDs on apoptosis and on the serine/threonine kinase pathway, Akt, and glycogen synthase kinase 3β (GSK3β). METHODS LNCaP cells, a type of prostate cancer cells (derived from left supraclavicular lymph node of human prostrate carcinoma), were cultured in DMEM medium, and cell viability was evaluated with a colorimetric assay using MTT level. The total and phosphorylated protein level of Akt and GSK3β were detected by Western blotting. RESULTS AND DISCUSSION The apoptosis-inducing effect of TZDs on prostate cancer cells involves the inhibition of Akt phosphorylation. Furthermore, TZDs induce inactivation of GSK3β, a multifunctional kinase that mediates essential events promoting prostate cancer development and acquisition of androgen independence. In addition, the GSK3β inhibitor lithium chloride sensitizes prostate cancer cells to TZDs cytotoxicity. WHAT IS NEW AND CONCLUSION Our data suggest that modulation of Akt-GSK3β pathway is involved in the cell death pathway engaged by TZDs in prostate cancer cells. This reveals another possible mechanism of TZDs on apoptosis in prostate cancer. Inhibition of the Akt-GSK3β cascade may be a useful approach in prostate cancer.
Collapse
Affiliation(s)
- H Zhu
- Department of Pathophysiology, Medical College of Qingdao University, Qingdao University, Qingdao, China
| | | | | | | | | |
Collapse
|
126
|
Freudlsperger C, Dahl A, Hoffmann J, Reinert S, Schumacher U. Mistletoe lectin-I augments antiproliferative effects of the PPARgamma agonist rosiglitazone on human malignant melanoma cells. Phytother Res 2011; 24:1354-8. [PMID: 20812278 DOI: 10.1002/ptr.3122] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
As malignant melanoma cells are highly resistant to conventional chemotherapy, survival rates after tumor spread remain poor and hence there is an urgent need for new therapeutic options. For both mistletoe lectin-I (ML-I) and the thiazolidinediones as synthetic ligands of the peroxisome proliferator-activated receptor gamma (PPARgamma) an antiproliferative effect on malignant melanoma cells has previously been shown. Hence, the aim of this study was to investigate whether the combination of ML-I and the PPARgamma ligand rosiglitazone is more efficacious in the treatment of malignant melanoma cells than either agent alone. Proliferation of three human melanoma cell lines treated with ML-I, rosiglitazone and the combination of both was measured in a broad concentration range (0.0001-100 microg/mL) using the XTT cell proliferation assay. Combined application tremendously increased the antiproliferative effect on all three melanoma cell lines compared with single agent treatment. In comparison with the single use of rosiglitazone, the combination with ML-I significantly increased the inhibition of cell growth by 51-79% and in comparison with the single use of ML-I by 9-32%, respectively. In conclusion, this study shows that the combination of ML-I with rosiglitazone significantly augments their antiproliferative effect on malignant melanoma cells in comparison with their single agent application, which might be a promising tool for further therapeutic studies.
Collapse
Affiliation(s)
- Christian Freudlsperger
- Institute of Anatomy II, University Hospital Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany.
| | | | | | | | | |
Collapse
|
127
|
Miao R, Xu T, Liu L, Wang M, Jiang Y, Li J, Guo R. Rosiglitazone and retinoic acid inhibit proliferation and induce apoptosis in the HCT-15 human colorectal cancer cell line. Exp Ther Med 2011; 2:413-417. [PMID: 22977519 DOI: 10.3892/etm.2011.227] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2010] [Accepted: 03/01/2011] [Indexed: 11/05/2022] Open
Abstract
The aim of this study was to explore the effects of rosiglitazone (RSG) in combination with all-trans retinoic acid (ATRA) on the proliferation and apoptosis of the HCT-15 human colorectal cancer cell line. HCT-15 cells were divided into a blank control group, a vehicle control group and experimental groups (RSG only or ATRA only or RSG plus ATRA). Growth inhibition was examined using the MTT assay. Apoptosis and cell cycle progression were examined by flow cytometry. The expression of COX-2, MMP-7 and TIMP-1 was examined by immunocytochemistry. RSG alone inhibited HCT-15 cell proliferation in a concentration- and time-dependent manner (P<0.05). The combination of RSG and ATRA exhibited significant synergy (q>1.15). RSG or ATRA alone effectively increased the proportion of cells in the G0/G1 phase and decreased the proportion of cells in the S phase, thus inducing apoptosis (P<0.05). The combination of RSG and ATRA resulted in even stronger G1 cell cycle arrest (P<0.05). HCT-15 cells expressed COX-2, MMP-7 and TIMP-1, with positive expression rates in the control group of 66.79, 73.21 and 64.08%, respectively. After the combined application of RSG and ATRA, the positive rates significantly declined to only 19.33, 20.58 and 13.13%, respectively (P<0.01). In conclusion, the combination of RSG and ATRA reduced the expression of COX-2, MMP-7 and TIMP-1, caused cell cycle arrest at the G1 phase and induced apoptosis, which resulted in the inhibition of cell proliferation in the HCT-15 human colorectal cancer cell line.
Collapse
|
128
|
Yang CM, Lu IH, Chen HY, Hu ML. Lycopene inhibits the proliferation of androgen-dependent human prostate tumor cells through activation of PPARγ-LXRα-ABCA1 pathway. J Nutr Biochem 2011; 23:8-17. [PMID: 21334870 DOI: 10.1016/j.jnutbio.2010.10.006] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2010] [Revised: 09/24/2010] [Accepted: 10/11/2010] [Indexed: 11/29/2022]
Abstract
The activation of nuclear receptors, peroxisome proliferator-activated receptor gamma (PPARγ) and liver X receptor alpha (LXRα), has been shown to inhibit the growth of prostate cancer cells. This study examined whether the anti-proliferative effect of lycopene on androgen-dependent human prostate cancer (LNCaP) cells involves the up-regulation of the expression of PPARγ and LXRα. As expected, lycopene treatment (2.5-10 μM) significantly inhibited the proliferation of LNCaP cells during incubation for 96 h. Lycopene significantly increased the protein and mRNA expression of PPARγ and LXRα at 24 and 48 h, while the increased in the expression of ATP-binding cassette transporter 1 (ABCA1) was only evident 96 h. In addition, lycopene significantly decreased cellular total cholesterol levels and increased apoA1 protein expression at 96 h. Incubation of LNCaP cells with lycopene (10 μM) in the presence (20 μM) of a specific antagonist of PPARγ (GW9662) and LXRα (GGPP) restored the proliferation of LNCaP cells to the control levels and significantly suppressed protein expression of PPARγ and LXRα as well as increased cellular total cholesterol levels. LXRα knockdown by siRNA against LXRα significantly enhanced the proliferation of LNCaP cells, whereas si-LXRα knockdown followed by incubation with lycopene (10 μM) restored the proliferation to the control level. The present study is the first to demonstrate that the anti-proliferative effect of lycopene on LNCaP cells involves the activation of the PPARγ-LXRα-ABCA1 pathway, leading to reduced cellular total cholesterol levels.
Collapse
Affiliation(s)
- Chih-Min Yang
- Department of Food Science and Biotechnology, National Chung Hsing University, Taichung, Taiwan 402, ROC
| | | | | | | |
Collapse
|
129
|
Malaguarnera R, Belfiore A. The insulin receptor: a new target for cancer therapy. Front Endocrinol (Lausanne) 2011; 2:93. [PMID: 22654833 PMCID: PMC3356071 DOI: 10.3389/fendo.2011.00093] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2011] [Accepted: 11/19/2011] [Indexed: 12/16/2022] Open
Abstract
A large body of evidences have shown that both the IGF-I receptor (IGF-IR) and the insulin receptor (IR) play a role in cancer development and progression. In particular, IR overactivation by IGF-II is common in cancer cells, especially in dedifferentiated/stem-like cells. In spite of these findings, until very recently, only IGF-IR but not IR has been considered a target in cancer therapy. Although several preclinical studies have showed a good anti-cancer activity of selective anti-IGF-IR drugs, the results of the clinical first trials have been disappointing. In fact, only a small subset of malignant tumors has shown an objective response to these therapies. Development of resistance to anti-IGF-IR drugs may include upregulation of IR isoform A (IR-A) in cancer cells and its overactivation by increased secretion of autocrine IGF-II. These findings have led to the concept that co-targeting IR together with IGF-IR may increase therapy efficacy and prevent adaptive resistance to selective anti-IGF-IR drugs. IR blockade should be especially considered in tumors with high IR-A:IGF-IR ratio and high levels of autocrine IGF-II. Conversely, insulin sensitizers, which ameliorate insulin resistance associated with metabolic disorders and cancer treatments, may have important implications for cancer prevention and management. Only few drugs co-targeting the IR and IGF-IR are currently available. Ideally, future IR targeting strategies should be able to selectively inhibit the tumor promoting effects of IR without impairing its metabolic effects.
Collapse
Affiliation(s)
- Roberta Malaguarnera
- Endocrinology Unit, Department of Clinical and Experimental Medicine, University Magna Graecia of CatanzaroCatanzaro, Italy
| | - Antonino Belfiore
- Endocrinology Unit, Department of Clinical and Experimental Medicine, University Magna Graecia of CatanzaroCatanzaro, Italy
- *Correspondence: Antonino Belfiore, Endocrinology Unit, Department of Clinical and Experimental Medicine, University of Catanzaro, Campus Universitario, Viale Europa, località Germaneto, 88100 Catanzaro, Italy. e-mail:
| |
Collapse
|
130
|
Peroxisome proliferator-activated receptor gamma ligand-mediated apoptosis of hepatocellular carcinoma cells depends upon modulation of PI3Kinase pathway independent of Akt. J Mol Signal 2010; 5:20. [PMID: 21144036 PMCID: PMC3009957 DOI: 10.1186/1750-2187-5-20] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2010] [Accepted: 12/13/2010] [Indexed: 11/25/2022] Open
Abstract
Background Ligands of Peroxisome proliferator-activated receptor gamma (PPARγ) can inhibit growth and promote apoptosis in various cancer cells, and thus have the potential to be utilized as anticancer drugs. This potential however, has been seriously challenged by observations that they can lead to tumor promotion in some cancer models, possibly due to activation of different signaling mechanisms in various tumor environments. Elucidation of the specific signaling events that modulate PPARγ ligand-mediated events is thus critical to increase their efficacy. The studies described here were designed to elucidate the signaling pathway(s) that modulate the apoptotic potential of Troglitazone (TRG), an artificial PPARγ ligand in hepatocellular carcinoma (HCC) cells. Results Our results indicate that the apoptotic potential of TRG was regulated by the presence or absence of serum in the media. When added in serum-containing media, TRG inhibited proliferation and cyclin D1 expression, but was unable to induce any apoptosis. However, TRG's apoptotic potential was induced significantly when added in serum deficient media, as indicated by increased PARP and Caspase-3 cleavage and results from apoptosis assay. Furthermore, TRG-induced apoptosis in serum deficient media was associated with a dramatic reduction in PI3Kinase downstream target AktSer473 and FoxO1Thr24/FoxO3aThr32 phosphorylation. On the contrary, there was an increase of PI3K-induced AktSer473 and FoxO1Thr24/FoxO3aThr32 phosphorylation involving Pak, when TRG was added in serum-containing media. Pharmacological inhibition of PI3Kinase pathway with LY294002 inhibited Aktser473 phosphorylation and sensitized cells towards apoptosis in the presence of serum, indicating the involvement of PI3K in apoptosis resistance. Interestingly, pharmacological inhibition or siRNA-mediated knockdown of Akt or inhibition of Pak was unable to sensitize cells towards TRG-induced apoptosis in the presence of serum. Similarly, TRG was unable to induce apoptosis in the Akt1-KO, Akt1&2-KO MEFs in serum-containing media. Conclusion These studies indicate that TRG-induced apoptosis is modulated by PI3K pathway in a novel Akt-independent manner, which might contribute to its tumor promoting effects. Since PI3K activation is linked with various cancers, combination therapy utilizing TRG and PI3K inhibitors has the potential to not only increase the efficacy of TRG as a chemotherapeutic agent but also reduce its off target effects.
Collapse
|
131
|
Moss PE, Lyles BE, Stewart LV. The PPARγ ligand ciglitazone regulates androgen receptor activation differently in androgen-dependent versus androgen-independent human prostate cancer cells. Exp Cell Res 2010; 316:3478-88. [PMID: 20932825 PMCID: PMC2977976 DOI: 10.1016/j.yexcr.2010.09.015] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2010] [Revised: 09/27/2010] [Accepted: 09/28/2010] [Indexed: 11/19/2022]
Abstract
The androgen receptor (AR) regulates growth and progression of androgen-dependent as well as androgen-independent prostate cancer cells. Peroxisome proliferator-activated receptor gamma (PPARγ) agonists have been reported to reduce AR activation in androgen-dependent LNCaP prostate cancer cells. To determine whether PPARγ ligands are equally effective at inhibiting AR activity in androgen-independent prostate cancer, we examined the effect of the PPARγ ligands ciglitazone and rosiglitazone on C4-2 cells, an androgen- independent derivative of the LNCaP cell line. Luciferase-based reporter assays and Western blot analysis demonstrated that PPARγ ligand reduced dihydrotestosterone (DHT)-induced increases in AR activity in LNCaP cells. However, in C4-2 cells, these compounds increased DHT-induced AR driven luciferase activity. In addition, ciglitazone did not significantly alter DHT-mediated increases in prostate specific antigen (PSA) protein or mRNA levels within C4-2 cells. siRNA-based experiments demonstrated that the ciglitazone-induced regulation of AR activity observed in C4-2 cells was dependent on the presence of PPARγ. Furthermore, overexpression of the AR corepressor cyclin D1 inhibited the ability of ciglitazone to induce AR luciferase activity in C4-2 cells. Thus, our data suggest that both PPARγ and cyclin D1 levels influence the ability of ciglitazone to differentially regulate AR signaling in androgen-independent C4-2 prostate cancer cells.
Collapse
Affiliation(s)
- Patrice E. Moss
- Department of Biochemistry and Cancer Biology, Meharry Medical College, Nashville, TN 37208
| | - Besstina E. Lyles
- Department of Biochemistry and Cancer Biology, Meharry Medical College, Nashville, TN 37208
| | - LaMonica V. Stewart
- Department of Biochemistry and Cancer Biology, Meharry Medical College, Nashville, TN 37208
| |
Collapse
|
132
|
Abstract
Differentiation therapy with all-trans retinoic acid has been a very successful therapeutic strategy in acute promyelocytic leukemia (APL), but the value of differentiation therapy in acute myeloid leukemia (AML) remains to be determined. A number of current treatments, such as tyrosine kinase inhibitors, cytokines, and epigenetic agents, induce differentiation of leukemic cells to some extent, but differentiation is not the main goal of these treatments. Forcing expression of certain transcription factors, such as C/EBP, has also been useful in inducing differentiation in cell lines and in murine models, but an effective way to force expression of these genes in humans is yet to be discovered.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents/therapeutic use
- CCAAT-Enhancer-Binding Proteins/biosynthesis
- CCAAT-Enhancer-Binding Proteins/genetics
- Cell Differentiation/drug effects
- Cell Differentiation/genetics
- Cell Line, Tumor
- Cricetinae
- Disease Models, Animal
- Gene Expression Regulation, Leukemic/drug effects
- Gene Expression Regulation, Leukemic/genetics
- Humans
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/therapy
- Leukemia, Promyelocytic, Acute
Collapse
Affiliation(s)
- H Phillip Koeffler
- Cedars-Sinai Medical Center, University of California Los Angeles School of Medicine, 90048, USA.
| |
Collapse
|
133
|
Galli A, Ceni E, Mello T, Polvani S, Tarocchi M, Buccoliero F, Lisi F, Cioni L, Ottanelli B, Foresta V, Mastrobuoni G, Moneti G, Pieraccini G, Surrenti C, Milani S. Thiazolidinediones inhibit hepatocarcinogenesis in hepatitis B virus-transgenic mice by peroxisome proliferator-activated receptor gamma-independent regulation of nucleophosmin. Hepatology 2010; 52:493-505. [PMID: 20683949 DOI: 10.1002/hep.23669] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
UNLABELLED Antidiabetic thiazolidinediones (TZD) have in vitro antiproliferative effect in epithelial cancers, including hepatocellular carcinoma (HCC). The effective anticancer properties and the underlying molecular mechanisms of these drugs in vivo remain unclear. In addition, the primary biological target of TZD, the ligand-dependent transcription factor peroxisome proliferator-activated receptor gamma (PPARgamma), is up-regulated in HCC and seems to provide tumor-promoting responses. The aim of our study was to evaluate whether chronic administration of TZD may affect hepatic carcinogenesis in vivo in relation to PPARgamma expression and activity. The effect of TZD oral administration for 26 weeks was tested on tumor formation in PPARgamma-expressing and PPARgamma-deficient mouse models of hepatic carcinogenesis. Proteomic analysis was performed in freshly isolated hepatocytes by differential in gel electrophoresis and mass spectrometry analysis. Identified TZD targets were confirmed in cultured PPARgamma-deficient hepatocytes. TZD administration in hepatitis B virus (HBV)-transgenic mice (TgN[Alb1HBV]44Bri) reduced tumor incidence in the liver, inhibiting hepatocyte proliferation and increasing apoptosis. PPARgamma deletion in hepatocytes of HBV-transgenic mice (Tg[HBV]CreKOgamma) did not modify hepatic carcinogenesis but increased the TZD antitumorigenic effect. Proteomic analysis identified nucleophosmin (NPM) as a TZD target in PPARgamma-deficient hepatocytes. TZD inhibited NPM expression at protein and messenger RNA levels and decreased NPM promoter activity. TZD inhibition of NPM was associated with the induction of p53 phosphorylation and p21 expression. CONCLUSION These findings suggest that chronic administration of TZD has anticancer activity in the liver via inhibition of NPM expression and indicate that these drugs might be useful for HCC chemoprevention and treatment.
Collapse
Affiliation(s)
- Andrea Galli
- Gastroenterology Unit, Department of Clinical Pathophysiology, University of Florence, Florence, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
134
|
Rashid-Kolvear F, Taboski MAS, Nguyen J, Wang DY, Harrington LA, Done SJ. Troglitazone suppresses telomerase activity independently of PPARgamma in estrogen-receptor negative breast cancer cells. BMC Cancer 2010; 10:390. [PMID: 20650001 PMCID: PMC2915983 DOI: 10.1186/1471-2407-10-390] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2009] [Accepted: 07/22/2010] [Indexed: 12/03/2022] Open
Abstract
Background Breast cancer is one the highest causes of female cancer death worldwide. Many standard chemotherapeutic agents currently used to treat breast cancer are relatively non-specific and act on all rapidly dividing cells. In recent years, more specific targeted therapies have been introduced. It is known that telomerase is active in over 90% of breast cancer tumors but inactive in adjacent normal tissues. The prevalence of active telomerase in breast cancer patients makes telomerase an attractive therapeutic target. Recent evidence suggests that telomerase activity can be suppressed by peroxisome proliferator activated receptor gamma (PPARγ). However, its effect on telomerase regulation in breast cancer has not been investigated. Methods In this study, we investigated the effect of the PPARγ ligand, troglitazone, on telomerase activity in the MDA-MB-231 breast cancer cell line. Real time RT-PCR and telomerase activity assays were used to evaluate the effect of troglitazone. MDA-MB-231 cells had PPARγ expression silenced using shRNA interference. Results We demonstrated that troglitazone reduced the mRNA expression of hTERT and telomerase activity in the MDA-MB-231 breast cancer cell line. Troglitazone reduced telomerase activity even in the absence of PPARγ. In agreement with this result, we found no correlation between PPARγ and hTERT mRNA transcript levels in breast cancer patients. Statistical significance was determined using Pearson correlation and the paired Student's t test. Conclusions To our knowledge, this is the first time that the effect of troglitazone on telomerase activity in breast cancer cells has been investigated. Our data suggest that troglitazone may be used as an anti-telomerase agent; however, the mechanism underlying this inhibitory effect remains to be determined.
Collapse
Affiliation(s)
- Fariborz Rashid-Kolvear
- Department of Laboratory Medicine and Pathobiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | | | | | | | | | | |
Collapse
|
135
|
Holbeck S, Chang J, Best AM, Bookout AL, Mangelsdorf DJ, Martinez ED. Expression profiling of nuclear receptors in the NCI60 cancer cell panel reveals receptor-drug and receptor-gene interactions. Mol Endocrinol 2010; 24:1287-96. [PMID: 20375240 PMCID: PMC2875807 DOI: 10.1210/me.2010-0040] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2010] [Accepted: 03/17/2010] [Indexed: 12/15/2022] Open
Abstract
We profiled the expression of the 48 human nuclear receptors (NRs) by quantitative RT-PCR in 51 human cancer cell lines of the NCI60 collection derived from nine different tissues. NR mRNA expression accurately classified melanoma, colon, and renal cancers, whereas lung, breast, prostate, central nervous system, and leukemia cell lines exhibited heterogeneous receptor expression. Importantly, receptor mRNA levels faithfully predicted the growth-inhibitory qualities of receptor ligands in nonendocrine tumors. Correlation analysis using NR expression profiles and drug response information across the cell line panel uncovered a number of new potential receptor-drug interactions, suggesting that in these cases, individual receptor levels may predict response to chemotherapeutic interventions. Similarly, by cross-comparing receptor levels within our expression dataset and relating these profiles to existing microarray gene expression data, we defined interactions among receptors and between receptors and other genes that can now be mechanistically queried. This work supports the strategy of using NR expression profiling to classify various types of cancer, define NR-drug interactions and receptor-gene networks, predict cancer-drug sensitivity, and identify druggable targets that may be pharmacologically manipulated for potential therapeutic intervention.
Collapse
Affiliation(s)
- Susan Holbeck
- National Cancer Institute, National Institutes of Health, Rockville, Maryland 20852, USA
| | | | | | | | | | | |
Collapse
|
136
|
Pioglitazone modulates tumor cell metabolism and proliferation in multicellular tumor spheroids. Cancer Chemother Pharmacol 2010; 67:117-26. [PMID: 20217088 DOI: 10.1007/s00280-010-1294-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2009] [Accepted: 02/13/2010] [Indexed: 10/19/2022]
Abstract
The anti-diabetic thiazolidinedione compound pioglitazone, a peroxisome proliferator-activated receptor-gamma agonist, and selective cyclooxygenase-2 inhibitors are clinically used in patients with advanced malignancies. Several previously published in vivo and in vitro studies showed growth inhibitory effects on different cancer cell lines. However, the underlying mechanisms are fairly unclear. Here, we analyzed the effects of pioglitazone in combination with other drugs in a three-dimensional multicellular tumor spheroid culture system (MCTS) generated from the two prostate carcinoma cell lines PC3 and LNCaP. As expected, pioglitazone also inhibited tumor cell proliferation in the MCTS system. Further studies revealed that pioglitazone lowered the pH of the culture medium, decreased oxygen consumption and increased lactate secretion in both tumor cell lines. Other glitazones, troglitazone and ciglitazone, had similar effects. The combination of pioglitazone with 2-deoxyglucose, a potent inhibitor of glycolysis, had an additive effect on the inhibition of cell proliferation and led to MCTS disintegration. Our data propose a new mechanism of growth inhibition by pioglitazone through modulation of the tumor cell metabolism.
Collapse
|
137
|
Knopfová L, Smarda J. The use of Cox-2 and PPARγ signaling in anti-cancer therapies. Exp Ther Med 2010; 1:257-264. [PMID: 22993537 DOI: 10.3892/etm_00000040] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2009] [Accepted: 11/02/2009] [Indexed: 02/06/2023] Open
Abstract
Increased production of the pro-inflammatory enzyme cyclooxygenase-2 (Cox-2) and altered expression and activity of peroxisome proliferator-activated receptor γ (PPARγ) have been observed in many malignancies. Both the PPARγ ligands and the Cox-2 inhibitors possess anti-inflammatory and anti-neoplastic effects in vitro and have been assessed for their therapeutic potential in several pre-clinical and clinical studies. Recently, multiple interactions between PPARγ and Cox-2 signaling pathways have been revealed. Understanding of the cross-talk between PPARγ and Cox-2 might provide important novel strategies for the effective treatment and/or prevention of cancer. This article summarizes recent achievements involving the functional interactions between the PPARγ and Cox-2 signaling pathways and discusses the implications of such interplay for clinical use.
Collapse
Affiliation(s)
- Lucia Knopfová
- Department of Experimental Biology, Faculty of Science, Masaryk University, 611 37 Brno, Czech Republic
| | | |
Collapse
|
138
|
Davies G, Ross A, Arnason T, Juurlink B, Harkness T. Troglitazone inhibits histone deacetylase activity in breast cancer cells. Cancer Lett 2010; 288:236-50. [DOI: 10.1016/j.canlet.2009.07.011] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2009] [Revised: 07/09/2009] [Accepted: 07/10/2009] [Indexed: 10/20/2022]
|
139
|
Differential effects of PPARgamma activation by the oral antidiabetic agent pioglitazone in Barrett's carcinoma in vitro and in vivo. J Gastroenterol 2010; 44:919-29. [PMID: 19506796 DOI: 10.1007/s00535-009-0086-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2008] [Accepted: 05/11/2009] [Indexed: 02/04/2023]
Abstract
BACKGROUND AND PURPOSE The nuclear hormone receptor peroxisome proliferator-activated receptor gamma (PPARgamma) is a key transcription factor regulating genes involved in adipogenesis, glucose homeostasis and cell differentiation. Moreover, PPARgamma has been demonstrated to control proliferation and apoptosis in various cancer cells. We investigated the biological effects of PPARgamma activation by the oral antidiabetic agent pioglitazone in Barrett's adenocarcinoma cells in vitro and in vivo. RESULTS PPARgamma mRNA and protein were overexpressed in endoscopic biopsies of Barrett's epithelium and the human Barrett's adenocarcinoma cancer cell line OE33 as compared to normal esophagus and stomach and the esophageal squamous epithelium cancer cell line Kyse-180. PPARgamma activation by pioglitazone in OE33 cells in vitro led to reduced cell growth by induction of apoptosis. Effects of systemic PPARgamma activation by the thiazolidinedione pioglitazone on tumor cell proliferation and apoptosis were then assessed in vivo in nude mice bearing transplantable Barrett's adenocarcinomas derived from OE33 cells. Unexpectedly, enhanced growth of OE33 derived transplantable adenocarcinomas was observed in Balb/c nu/nu mice upon systemic pioglitazone treatment due to increased cell proliferation. CONCLUSION These results indicate that PPARgamma is involved in the molecular pathogenesis of Barrett's adenocarcinoma formation and growth. However, activation of PPARgamma exerts differential effects on growth of Barrett's adenocarcinoma cells in vitro and in vivo emphasizing the importance of additional cell context specific factors and systemic metabolic status for the modulation of PPARgamma action in vivo.
Collapse
|
140
|
Cellai I, Petrangolini G, Tortoreto M, Pratesi G, Luciani P, Deledda C, Benvenuti S, Ricordati C, Gelmini S, Ceni E, Galli A, Balzi M, Faraoni P, Serio M, Peri A. In vivo effects of rosiglitazone in a human neuroblastoma xenograft. Br J Cancer 2010; 102:685-92. [PMID: 20068562 PMCID: PMC2837558 DOI: 10.1038/sj.bjc.6605506] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Neuroblastoma (NB) is the most common extra-cranial solid tumour in infants. Unfortunately, most children present with advanced disease and have a poor prognosis. There is in vitro evidence that the peroxisome proliferator-activated receptor gamma (PPARgamma) might be a target for pharmacological intervention in NB. We have previously demonstrated that the PPARgamma agonist rosiglitazone (RGZ) exerts strong anti-tumoural effects in the human NB cell line, SK-N-AS. The aim of this study was to evaluate whether RGZ maintains its anti-tumoural effects against SK-N-AS NB cells in vivo. METHODS AND RESULTS For this purpose, tumour cells were subcutaneously implanted in nude mice, and RGZ (150 mg kg(-1)) was administered by gavage daily for 4 weeks. At the end of treatment, a significant tumour weight inhibition (70%) was observed in RGZ-treated mice compared with control mice. The inhibition of tumour growth was supported by a strong anti-angiogenic activity, as assessed by CD-31 immunostaining in tumour samples. The number of apoptotic cells, as determined by cleaved caspase-3 immunostaining, seemed lower in RGZ-treated animals at the end of the treatment period than in control mice, likely because of the large tumour size observed in the latter group. CONCLUSIONS To our knowledge, this is the first demonstration that RGZ effectively inhibits tumour growth in a human NB xenograft and our results suggest that PPARgamma agonists may have a role in anti-tumoural strategies against NB.
Collapse
Affiliation(s)
- I Cellai
- Department of Clinical Physiopathology, Center for Research, University of Florence, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
141
|
Hsieh WC, Lan BS, Chen YL, Chang Y, Chuang HC, Su IJ. Efficacy of peroxisome proliferator activated receptor agonist in the treatment of virus-associated haemophagocytic syndrome in a rabbit model. Antivir Ther 2010; 15:71-81. [DOI: 10.3851/imp1490] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
142
|
Venkatachalam G, Kumar AP, Yue LS, Pervaiz S, Clement MV, Sakharkar MK. Computational identification and experimental validation of PPRE motifs in NHE1 and MnSOD genes of human. BMC Genomics 2009; 10 Suppl 3:S5. [PMID: 19958503 PMCID: PMC2788392 DOI: 10.1186/1471-2164-10-s3-s5] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Activation of PPARs has been reported to inhibit the proliferation of malignant cells from different lineages. They are involved in transcription regulation of genes upon activation by a ligand. The binding of PPARs to the promoter sequence either represses or activates the gene. Hence, PPARs represent promising targets for cancer treatment because of their anti-proliferative and pro-apoptotic activities. Here we computationally identified PPAR binding regions in NHE1 and MnSOD. We further validated the predictions in vitro. RESULTS Our results computationally predicted the presence of 2 PPRE motifs in NHE1 and 3 PPRE motifs in MnSOD. We experimentally confirmed the true motifs and their regulation by PPAR. CONCLUSION Our results suggest that both NHE1 and MnSOD have PPRE binding motif in their upstream/promoter region and hence are regulated by PPAR upon ligand binding.
Collapse
Affiliation(s)
- Gireedhar Venkatachalam
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
| | | | | | | | | | | |
Collapse
|
143
|
Zabirnyk O, Liu W, Khalil S, Sharma A, Phang JM. Oxidized low-density lipoproteins upregulate proline oxidase to initiate ROS-dependent autophagy. Carcinogenesis 2009; 31:446-54. [PMID: 19942609 PMCID: PMC2832543 DOI: 10.1093/carcin/bgp299] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Epidemiological studies showed that high levels of oxidized low-density lipoproteins (oxLDLs) are associated with increased cancer risk. We examined the direct effect of physiologic concentrations oxLDL on cancer cells. OxLDLs were cytotoxic and activate both apoptosis and autophagy. OxLDLs have ligands for peroxisome proliferator-activated receptor gamma and upregulated proline oxidase (POX) through this nuclear receptor. We identified 7-ketocholesterol (7KC) as a main component responsible for the latter. To elucidate the role of POX in oxLDL-mediated cytotoxicity, we knocked down POX via small interfering RNA and found that this (i) further reduced viability of cancer cells treated with oxLDL; (ii) decreased oxLDL-associated reactive oxygen species generation; (iii) decreased autophagy measured via beclin-1 protein level and light-chain 3 protein (LC3)-I into LC3-II conversion. Using POX-expressing cell model, we established that single POX overexpression was sufficient to activate autophagy. Thus, it led to autophagosomes accumulation and increased conversion of LC3-I into LC3-II. Moreover, beclin-1 gene expression was directly dependent on POX catalytic activity, namely the generation of POX-dependent superoxide. We conclude that POX is critical in the cellular response to the noxious effects of oxLDL by activating protective autophagy.
Collapse
Affiliation(s)
- Olga Zabirnyk
- Metabolism and Cancer Susceptibility Section, Laboratory of Comparative Carcinogenesis, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702-1201, USA
| | | | | | | | | |
Collapse
|
144
|
Jiang M, Fernandez S, Jerome WG, He Y, Yu X, Cai H, Boone B, Yi Y, Magnuson MA, Roy-Burman P, Matusik RJ, Shappell SB, Hayward SW. Disruption of PPARgamma signaling results in mouse prostatic intraepithelial neoplasia involving active autophagy. Cell Death Differ 2009; 17:469-81. [PMID: 19834493 PMCID: PMC2821953 DOI: 10.1038/cdd.2009.148] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Peroxisome proliferator-activated receptor-gamma (PPARgamma) regulates the interface between cellular lipid metabolism, redox status and organelle differentiation. Conditional prostatic epithelial knockout of PPARgamma in mice resulted in focal hyperplasia which developed into mouse prostatic intraepithelial neoplasia (mPIN). The grade of PIN became more severe with time. Electron microscopy (EM) showed accumulated secondary lysosomes containing cellular organelles and debris suggestive of autophagy. Consistent with this analysis the autophagy marker LC-3 was found to be upregulated in areas of PIN in PPARgamma KO tissues. We selectively knocked down PPARgamma2 isoform in wild-type mouse prostatic epithelial cells and examined the consequences of this in a tissue recombination model. Histopathologically grafted tissues resembled the conditional PPARgamma KO mouse prostates. EM studies of PPARgamma- and PPARgamma2-deficient epithelial cells in vitro were suggestive of autophagy, consistent with the prostatic tissue analysis. This was confirmed by examining expression of beclin-1 and LC-3. Gene expression profiling in PPARgamma-/gamma2-deficient cells indicated a major dysregulation of cell cycle control and metabolic signaling networks related to peroxisomal and lysosomal maturation, lipid oxidation and degradation. The putative autophagic phenotypes of PPARgamma-deficient cells could be rescued by re-expression of either gamma1 or gamma2 isoform. We conclude that disruption of PPARgamma signaling results in autophagy and oxidative stress during mPIN pathogenesis.
Collapse
Affiliation(s)
- M Jiang
- Department of Urologic Surgery, A-1302 MCN, Vanderbilt University Medical Center, Nashville, TN 37232-2765, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
145
|
Phase II Study of Nitric Oxide Donor for Men With Increasing Prostate-specific Antigen Level After Surgery or Radiotherapy for Prostate Cancer. Urology 2009; 74:878-83. [DOI: 10.1016/j.urology.2009.03.004] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2009] [Revised: 02/24/2009] [Accepted: 03/02/2009] [Indexed: 11/23/2022]
|
146
|
van Kruijsdijk RCM, van der Wall E, Visseren FLJ. Obesity and cancer: the role of dysfunctional adipose tissue. Cancer Epidemiol Biomarkers Prev 2009; 18:2569-78. [PMID: 19755644 DOI: 10.1158/1055-9965.epi-09-0372] [Citation(s) in RCA: 517] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Overweight and obesity are health problems of epidemic proportions, increasing the risk not only of cardiovascular disease and type 2 diabetes mellitus but also of various types of cancer. Obesity is strongly associated with changes in the physiological function of adipose tissue, leading to insulin resistance, chronic inflammation, and altered secretion of adipokines. Several of these factors, such as insulin resistance, increased levels of leptin, plasminogen activator inhibitor-1, and endogenous sex steroids, decreased levels of adiponectin, and chronic inflammation, are involved in carcinogenesis and cancer progression. This article reviews these mechanisms, focusing on adipose tissue dysfunction as a unifying causal factor. Although understanding of the link between obesity and cancer might provide therapeutic targets, preventing overweight and obesity still remains number one priority.
Collapse
Affiliation(s)
- Rob C M van Kruijsdijk
- Department of Vascular Medicine, University Medical Center Utrecht, PO Box 85500, 3508 GA Utrecht, The Netherlands
| | | | | |
Collapse
|
147
|
Mo WJ, Fu XP, Han XT, Yang GY, Zhang JG, Guo FH, Huang Y, Mao YM, Li Y, Xie Y. A stochastic model for identifying differential gene pair co-expression patterns in prostate cancer progression. BMC Genomics 2009; 10:340. [PMID: 19640296 PMCID: PMC2737000 DOI: 10.1186/1471-2164-10-340] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2009] [Accepted: 07/29/2009] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The identification of gene differential co-expression patterns between cancer stages is a newly developing method to reveal the underlying molecular mechanisms of carcinogenesis. Most researches of this subject lack an algorithm useful for performing a statistical significance assessment involving cancer progression. Lacking this specific algorithm is apparently absent in identifying precise gene pairs correlating to cancer progression. RESULTS In this investigation we studied gene pair co-expression change by using a stochastic process model for approximating the underlying dynamic procedure of the co-expression change during cancer progression. Also, we presented a novel analytical method named 'Stochastic process model for Identifying differentially co-expressed Gene pair' (SIG method). This method has been applied to two well known prostate cancer data sets: hormone sensitive versus hormone resistant, and healthy versus cancerous. From these data sets, 428,582 gene pairs and 303,992 gene pairs were identified respectively. Afterwards, we used two different current statistical methods to the same data sets, which were developed to identify gene pair differential co-expression and did not consider cancer progression in algorithm. We then compared these results from three different perspectives: progression analysis, gene pair identification effectiveness analysis, and pathway enrichment analysis. Statistical methods were used to quantify the quality and performance of these different perspectives. They included: Re-identification Scale (RS) and Progression Score (PS) in progression analysis, True Positive Rate (TPR) in gene pair analysis, and Pathway Enrichment Score (PES) in pathway analysis. Our results show small values of RS and large values of PS, TPR, and PES; thus, suggesting that gene pairs identified by the SIG method are highly correlated with cancer progression, and highly enriched in disease-specific pathways. From this research, several gene interaction networks inferred could provide clues for the mechanism of prostate cancer progression. CONCLUSION The SIG method reliably identifies cancer progression correlated gene pairs, and performs well both in gene pair ontology analysis and in pathway enrichment analysis. This method provides an effective means of understanding the molecular mechanism of carcinogenesis by appropriately tracking down the process of cancer progression.
Collapse
Affiliation(s)
- Wen Juan Mo
- State Key Laboratory of Genetic Engineering, Institute of Genetics, School of Life Science, Fudan University, Shanghai 200433, PR China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
148
|
Meyer S, Vogt T, Landthaler M, Berand A, Reichle A, Bataille F, Marx AH, Menz A, Hartmann A, Kunz-Schughart LA, Wild PJ. Cyclooxygenase 2 (COX2) and Peroxisome Proliferator-Activated Receptor Gamma (PPARG) Are Stage-Dependent Prognostic Markers of Malignant Melanoma. PPAR Res 2009; 2009:848645. [PMID: 19639032 PMCID: PMC2712952 DOI: 10.1155/2010/848645] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2009] [Accepted: 04/14/2009] [Indexed: 11/17/2022] Open
Abstract
Using tissue microarrays (TMAs) we studied COX2/PPARG immunoreactivity in a broad spectrum of tumors focussing on clinicopathological correlations and the outcome of patients with malignant melanoma (MM). TMA-1 contained normal and tumor tissues (n = 3448) from 47 organs including skin neoplasms (n = 323); TMA-2 88 primary MM, 101 metastases, and 161 benign nevi. Based on a biomodulatory approach combining COX/PPAR-targeting with metronomic low-dose chemotherapy metastases of 36 patients participating in a randomized trial with metastatic (stage IV) melanoma were investigated using TMA-3. COX2/PPARG immunoreactivity significantly increased from nevi to primary MM and metastases; COX2 positivity was associated with advanced Clark levels and shorter recurrence-free survival. Patients with PPARG-positive metastases and biomodulatory metronomic chemotherapy alone or combined with COX2/PPARG-targeting showed a significantly prolonged progression-free survival. Regarding primary MM, COX2 expression indicates an increased risk of tumor recurrence. In metastatic MM, PPARG expression may be a predicitive marker for response to biomodulatory stroma-targeted therapy.
Collapse
Affiliation(s)
- Stefanie Meyer
- Department of Dermatology, University of Regensburg, 93042 Regensburg, Germany
| | - Thomas Vogt
- Department of Dermatology, University of Regensburg, 93042 Regensburg, Germany
| | - Michael Landthaler
- Department of Dermatology, University of Regensburg, 93042 Regensburg, Germany
| | - Anna Berand
- Department of Hematology and Oncology, University of Regensburg, 93042 Regensburg, Germany
| | - Albrecht Reichle
- Department of Hematology and Oncology, University of Regensburg, 93042 Regensburg, Germany
| | - Frauke Bataille
- Institute of Pathology, University of Regensburg, 93042 Regensburg, Germany
| | - Andreas H. Marx
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Anne Menz
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Arndt Hartmann
- Institute of Clinical Pathology, University of Erlangen, 91054 Erlangen, Germany
| | | | - Peter J. Wild
- Institute of Surgical Pathology, University Hospital Zurich, Schmelzbergstrasse 12, 8091 Zurich, Switzerland
| |
Collapse
|
149
|
Nakamura Y, Suzuki T, Sugawara A, Arai Y, Sasano H. Peroxisome proliferator-activated receptor gamma in human prostate carcinoma. Pathol Int 2009; 59:288-93. [PMID: 19432669 DOI: 10.1111/j.1440-1827.2009.02367.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Peroxisome proliferator-activated receptor (PPAR) is a member of the nuclear hormone receptor superfamily of transcription factors. Peroxisome proliferator-activated receptor gamma (PPARgamma) plays an important role in the regulation of lipid homeostasis, adipogenesis, insulin resistance, and development of various organs. Agonists of PPARgamma have been also reported to inhibit proliferation of prostate carcinoma cells as in other human malignancies, and these synthetic ligands have been used in differentiation-mediated therapy of various human carcinomas associated with high levels of PPARgamma. The significance of PPARgamma expression, however, was unknown in human prostate carcinoma tissues. The purpose of the present study was therefore to examine the immunolocalization of PPARgamma in human prostate cancer tissues (40 cases) and correlate the findings with clinicopathological features of the patients in order to evaluate its possible biological significance. Twenty-nine patients were positive for PPARgamma immunoreactivity (73%) and a significant inverse correlation was detected between PPARgamma immunoreactivity, pT stage (P = 0.036), and serum concentration of prostate-specific antigen (P = 0.0004). In conclusion, PPARgamma immunoreactivity is considered to be a new clinicopathological parameter of human prostate cancer.
Collapse
Affiliation(s)
- Yasuhiro Nakamura
- Department of Pathology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | | | | | | | | |
Collapse
|
150
|
Belfiore A, Genua M, Malaguarnera R. PPAR-γ agonists and their effects on IGF-I receptor signaling: Implications for cancer. PPAR Res 2009; 2009:830501. [PMID: 19609453 PMCID: PMC2709717 DOI: 10.1155/2009/830501] [Citation(s) in RCA: 87] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2009] [Accepted: 05/04/2009] [Indexed: 01/04/2023] Open
Abstract
It is now well established that the development and progression of a variety of human malignancies are associated with dysregulated activity of the insulin-like growth factor (IGF) system. In this regard, promising drugs have been developed to target the IGF-I receptor or its ligands. These therapies are limited by the development of insulin resistance and compensatory hyperinsulinemia, which in turn, may stimulate cancer growth. Novel therapeutic approaches are, therefore, required. Synthetic PPAR-γ agonists, such as thiazolidinediones (TZDs), are drugs universally used as antidiabetic agents in patients with type 2 diabetes. In addition of acting as insulin sensitizers, PPAR-γ agonists mediate in vitro and in vivo pleiotropic anticancer effects. At least some of these effects appear to be linked with the downregulation of the IGF system, which is induced by the cross-talk of PPAR-γ agonists with multiple components of the IGF system signaling. As hyperinsulinemia is an emerging cancer risk factor, the insulin lowering action of PPAR-γ agonists may be expected to be also beneficial to reduce cancer development and/or progression. In light of these evidences, TZDs or other PPAR-γ agonists may be exploited in those tumors "addicted" to the IGF signaling and/or in tumors occurring in hyperinsulinemic patients.
Collapse
Affiliation(s)
- A Belfiore
- Endocrinology Unit, Department of Clinical and Experimental Medicine, University of Catanzaro, 88100 Catanzaro, Italy.
| | | | | |
Collapse
|