Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 137
Filter
1.
Biomolecules ; 14(6)2024 May 21.
Article in English | MEDLINE | ID: mdl-38927010

ABSTRACT

Nuclear hormone receptors exist in dynamic equilibrium between transcriptionally active and inactive complexes dependent on interactions with ligands, proteins, and chromatin. The present studies examined the hypothesis that endogenous ligands activate peroxisome proliferator-activated receptor-ß/δ (PPARß/δ) in keratinocytes. The phorbol ester treatment or HRAS infection of primary keratinocytes increased fatty acids that were associated with enhanced PPARß/δ activity. Fatty acids caused PPARß/δ-dependent increases in chromatin occupancy and the expression of angiopoietin-like protein 4 (Angptl4) mRNA. Analyses demonstrated that stearoyl Co-A desaturase 1 (Scd1) mediates an increase in intracellular monounsaturated fatty acids in keratinocytes that act as PPARß/δ ligands. The activation of PPARß/δ with palmitoleic or oleic acid causes arrest at the G2/M phase of the cell cycle of HRAS-expressing keratinocytes that is not found in similarly treated HRAS-expressing Pparb/d-null keratinocytes. HRAS-expressing Scd1-null mouse keratinocytes exhibit enhanced cell proliferation, an effect that is mitigated by treatment with palmitoleic or oleic acid. Consistent with these findings, the ligand activation of PPARß/δ with GW0742 or oleic acid prevented UVB-induced non-melanoma skin carcinogenesis, an effect that required PPARß/δ. The results from these studies demonstrate that PPARß/δ has endogenous roles in keratinocytes and can be activated by lipids found in diet and cellular components.


Subject(s)
Keratinocytes , PPAR delta , PPAR-beta , Stearoyl-CoA Desaturase , Keratinocytes/metabolism , Keratinocytes/drug effects , PPAR-beta/metabolism , PPAR-beta/genetics , Animals , Mice , Stearoyl-CoA Desaturase/metabolism , Stearoyl-CoA Desaturase/genetics , PPAR delta/metabolism , PPAR delta/genetics , Fatty Acids/metabolism , Angiopoietin-Like Protein 4/metabolism , Angiopoietin-Like Protein 4/genetics , Humans , Oleic Acid/pharmacology , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Fatty Acids, Monounsaturated/pharmacology , Fatty Acids, Monounsaturated/metabolism , Skin Neoplasms/metabolism , Skin Neoplasms/genetics , Skin Neoplasms/pathology
2.
Toxicol Sci ; 198(1): 2-3, 2024 Feb 28.
Article in English | MEDLINE | ID: mdl-38416075
3.
Metabolites ; 13(8)2023 Aug 10.
Article in English | MEDLINE | ID: mdl-37623879

ABSTRACT

Long-term ligand activation of PPARα in mice causes hepatocarcinogenesis through a mechanism that requires functional PPARα. However, hepatocarcinogenesis is diminished in both Ppara-null and PPARA-humanized mice, yet both lines develop age-related liver cancer independently of treatment with a PPARα agonist. Since PPARα is a master regulator of liver lipid metabolism in the liver, lipidomic analyses were carried out in wild-type, Ppara-null, and PPARA-humanized mice treated with and without the potent agonist GW7647. The levels of hepatic linoleic acid in Ppara-null and PPARA-humanized mice were markedly higher compared to wild-type controls, along with overall fatty liver. The number of liver CD4+ T cells was also lower in Ppara-null and PPARA-humanized mice and was negatively correlated with the elevated linoleic acid. Moreover, more senescent hepatocytes and lower serum TNFα and IFNγ levels were observed in Ppara-null and PPARA-humanized mice with age. These studies suggest a new role for PPARα in age-associated hepatocarcinogenesis due to altered lipid metabolism in Ppara-null and PPARA-humanized mice and the accumulation of linoleic acid as part of an overall fatty liver that is associated with loss of CD4+ T cells in the liver in both transgenic models. Since fatty liver is a known causal risk factor for liver cancer, Ppara-null and PPARA-humanized mice are valuable models for examining the mechanisms of PPARα and age-dependent hepatocarcinogenesis.

4.
Toxicol Sci ; 192(1): 1-2, 2023 03 20.
Article in English | MEDLINE | ID: mdl-36938612
5.
Arch Biochem Biophys ; 731: 109428, 2022 11 30.
Article in English | MEDLINE | ID: mdl-36228705

ABSTRACT

Cannabidiolic acid (CBDA) can activate peroxisome proliferator-activated receptor-α (PPARα) and PPARγ. Whether CBDA can activate PPARß/δ has not been examined sufficiently to date. Since previous studies showed that triple-negative breast cancer cells respond to activation of PPARß/δ, the present study examined the effect of CBDA in MDA-MB-231 cells and compared the activities of CBDA with known PPARß/δ agonists/antagonists. Expression of the PPARß/δ target genes angiopoietin-like 4 (ANGPTL4) and adipocyte differentiation-related protein (ADRP) was increased by CBDA. Interestingly, ligand activation of PPARß/δ with GW501516 caused an increase in expression of both ANGPTL4 and ADRP, but the magnitude of this effect was markedly increased when co-treated with CBDA. Specificity of these effects were confirmed by showing that CBDA-induced expression of ANGPTL4 and ADRP is mitigated in the presence of either a PPARß/δ antagonist or an inverse agonist. Results from these studies suggest that CBDA can synergize with PPARß/δ and might interact with endogenous agonists that modulate PPARß/δ function.


Subject(s)
Cannabinoids , PPAR delta , PPAR-beta , PPAR-beta/genetics , PPAR-beta/metabolism , PPAR delta/genetics , PPAR delta/metabolism , PPAR alpha
6.
Toxicol Sci ; 186(2): 177-178, 2022 03 28.
Article in English | MEDLINE | ID: mdl-35348797
7.
Toxicology ; 465: 153056, 2022 01 15.
Article in English | MEDLINE | ID: mdl-34861291

ABSTRACT

Perfluorooctane sulfonate (PFOS) is a stable environmental contaminant that can activate peroxisome proliferator-activated receptor alpha (PPARα). In the present work, the specific role of mouse and human PPARα in mediating the hepatic effects of PFOS was examined in short-term studies using wild type, Ppara-null and PPARA-humanized mice. Mice fed 0.006 % PFOS for seven days (∼10 mg/kg/day), or 0.003 % PFOS for twenty-eight days (∼5 mg/kg/day), exhibited higher liver and serum PFOS concentrations compared to controls. Relative liver weights were also higher following exposure to dietary PFOS in all three genotypes as compared vehicle fed control groups. Histopathological examination of liver sections from mice treated for twenty-eight days with 0.003 % PFOS revealed a phenotype consistent with peroxisome proliferation, in wild-type and PPARA-humanized mice that was not observed in Ppara-null mice. With both exposures, expression of the PPARα target genes, Acox1, Cyp4a10, was significantly increased in wild type mice but not in Ppara-null or PPARA-humanized mice. By contrast, expression of the constitutive androstane receptor (CAR) target gene, Cyp2b10, and the pregnane X receptor (PXR) target gene, Cyp3a11, were higher in response to PFOS administration in all three genotypes compared to controls for both exposure periods. These results indicate that mouse PPARα can be activated in the liver by PFOS causing increased expression of Acox1, Cyp4a10 and histopathological changes in the liver. While histopathological analyses indicated the presence of mouse PPARα-dependent hepatic peroxisome proliferation in wild-type (a response associated with activation of PPARα) and a similar phenotype in PPARA-humanized mice, the lack of increased Acox1 and Cyp4a10 mRNA by PFOS in PPARA-humanized mice indicates that the human PPARα was not as responsive to PFOS as mouse PPARα with this dose regimen. Moreover, results indicate that hepatomegaly caused by PFOS does not require mouse or human PPARα and could be due to effects induced by activation of CAR and/or PXR.


Subject(s)
Alkanesulfonic Acids/toxicity , Chemical and Drug Induced Liver Injury/etiology , Environmental Pollutants/toxicity , Fluorocarbons/toxicity , Liver/drug effects , PPAR alpha/agonists , Acyl-CoA Oxidase/genetics , Acyl-CoA Oxidase/metabolism , Animals , Aryl Hydrocarbon Hydroxylases/genetics , Aryl Hydrocarbon Hydroxylases/metabolism , Chemical and Drug Induced Liver Injury/genetics , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Constitutive Androstane Receptor/agonists , Constitutive Androstane Receptor/genetics , Constitutive Androstane Receptor/metabolism , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , Cytochrome P450 Family 2/genetics , Cytochrome P450 Family 2/metabolism , Dose-Response Relationship, Drug , Humans , Liver/metabolism , Liver/pathology , Male , Mice, 129 Strain , Mice, Knockout , PPAR alpha/genetics , PPAR alpha/metabolism , Pregnane X Receptor/agonists , Pregnane X Receptor/genetics , Pregnane X Receptor/metabolism , Signal Transduction , Species Specificity , Steroid Hydroxylases/genetics , Steroid Hydroxylases/metabolism
8.
Biol Pharm Bull ; 44(11): 1598-1606, 2021.
Article in English | MEDLINE | ID: mdl-34719638

ABSTRACT

Excessive, chronic alcohol consumption can lead to alcoholic liver disease. The etiology of alcoholic liver disease is multifactorial and is influenced by alterations in gene expression and changes in fatty acid metabolism, oxidative stress, and insulin resistance. These events can lead to steatosis, fibrosis, and eventually to cirrhosis and liver cancer. Many of these functions are regulated by peroxisome proliferator-activated receptors (PPARs). Thus, it is not surprising that PPARs can modulate the mechanisms that cause alcoholic liver disease. While the roles of PPARα and PPARγ are clearer, the role of PPARß/δ in alcoholic liver disease requires further clarification. This review summarizes the current understanding based on recent studies that indicate that PPARß/δ can likely be targeted for the treatment and/or the prevention of alcoholic liver disease.


Subject(s)
Liver Diseases, Alcoholic/prevention & control , PPAR gamma/drug effects , PPAR-beta/drug effects , Animals , Humans , Liver/drug effects , Liver/metabolism , Liver Diseases, Alcoholic/drug therapy
9.
Annu Rev Anal Chem (Palo Alto Calif) ; 14(1): 467-487, 2021 07 27.
Article in English | MEDLINE | ID: mdl-34314226

ABSTRACT

High-resolution mass spectrometry (MS) has advanced the study of metabolism in living systems by allowing many metabolites to be measured in a single experiment. Although improvements in mass detector sensitivity have facilitated the detection of greater numbers of analytes, compound identification strategies, feature reduction software, and data sharing have not kept up with the influx of MS data. Here, we discuss the ongoing challenges with MS-based metabolomics, including de novo metabolite identification from mass spectra, differentiation of metabolites from environmental contamination, chromatographic separation of isomers, and incomplete MS databases. Because of their popularity and sensitive detection of small molecules, this review focuses on the challenges of liquid chromatography-mass spectrometry-based methods. We then highlight important instrumentational, experimental, and computational tools that have been created to address these challenges and how they have enabled the advancement of metabolomics research.


Subject(s)
Metabolomics , Software , Chromatography, Liquid , Databases, Factual , Mass Spectrometry
10.
Toxicol Sci ; 183(1): 70-80, 2021 08 30.
Article in English | MEDLINE | ID: mdl-34081128

ABSTRACT

Ppara-null and PPARA-humanized mice are refractory to hepatocarcinogenesis caused by the peroxisome proliferator-activated receptor-α (PPARα) agonist Wy-14,643. However, the duration of these earlier studies was limited to approximately 1 year of treatment, and the ligand used has a higher affinity for the mouse PPARα compared to the human PPARα. Thus, the present study examined the effect of long-term administration of a potent, high-affinity human PPARα agonist (GW7647) on hepatocarcinogenesis in wild-type, Ppara-null, or PPARA-humanized mice. In wild-type mice, GW7647 caused hepatic expression of known PPARα target genes, hepatomegaly, hepatic MYC expression, hepatic cytotoxicity, and a high incidence of hepatocarcinogenesis. By contrast, these effects were essentially absent in Ppara-null mice or diminished in PPARA-humanized mice, although hepatocarcinogenesis was observed in both genotypes. Enhanced fatty change (steatosis) was also observed in both Ppara-null and PPARA-humanized mice independent of GW7647. PPARA-humanized mice administered GW7647 also exhibited increased necrosis after 5 weeks of treatment. Results from these studies demonstrate that the mouse PPARα is required for hepatocarcinogenesis induced by GW7647 administered throughout adulthood. Results also indicate that a species difference exists between rodents and human PPARα in the response to ligand activation of PPARα. The hepatocarcinogenesis observed in control and treated Ppara-null mice is likely mediated in part by increased hepatic fatty change, whereas the hepatocarcinogenesis observed in PPARA-humanized mice may also be due to enhanced fatty change and cytotoxicity that could be influenced by the minimal activity of the human PPARα in this mouse line on downstream mouse PPARα target genes. The Ppara-null and PPARA-humanized mouse models are valuable tools for examining the mechanisms of PPARα-induced hepatocarcinogenesis, but the background level of liver cancer must be controlled for in the design and interpretation of studies that use these mice.


Subject(s)
Fatty Liver , Liver Neoplasms , Adult , Animals , Humans , Liver Neoplasms/chemically induced , Liver Neoplasms/genetics , Mice , Mice, Knockout , PPAR alpha/genetics
11.
Toxicol Sci ; 183(1): 81-92, 2021 08 30.
Article in English | MEDLINE | ID: mdl-34081146

ABSTRACT

Evidence suggests that species differences exist between rodents and humans in their biological responses to ligand activation of PPARα. Moreover, neonatal/postnatal rodents may be more sensitive to the effects of activating PPARα. Thus, the present studies examined the effects of chronic ligand activation of PPARα initiated during early neonatal development and continued into adulthood on hepatocarcinogenesis in mice. Wild-type, Ppara-null, or PPARA-humanized mice were administered a potent, high-affinity human PPARα agonist GW7647, and cohorts of mice were examined over time. Activation of PPARα with GW7647 increased expression of known PPARα target genes in liver and was associated with hepatomegaly, increased hepatic cytotoxicity and necrosis, increased expression of hepatic MYC, and a high incidence of hepatocarcinogenesis in wild-type mice. These effects did not occur or were largely diminished in Ppara-null and PPARA-humanized mice, although background levels of hepatocarcinogenesis were also noted in both Ppara-null and PPARA-humanized mice. More fatty change (steatosis) was also observed in both Ppara-null and PPARA-humanized mice independent of GW7647 administration. Results from these studies indicate that the mouse PPARα is required to mediate hepatocarcinogenesis induced by GW7647 in mice and that activation of the human PPARα with GW7647 in PPARA-humanized mice are diminished compared with wild-type mice. Ppara-null and PPARA-humanized mice are valuable tools for examining species differences in the mechanisms of PPARα-induced hepatocarcinogenesis, but background levels of liver cancer observed in aged Ppara-null and PPARA-humanized mice must be considered when interpreting results from studies that use these models. These results also demonstrate that early life exposure to a potent human PPARα agonist does not enhance sensitivity to hepatocarcinogenesis.


Subject(s)
Fatty Liver , Liver Neoplasms , Adult , Aged , Animals , Female , Humans , Liver , Liver Neoplasms/chemically induced , Mice , Mice, Knockout , PPAR alpha/genetics , Pregnancy , Species Specificity
12.
13.
Toxicology ; 431: 152365, 2020 02 15.
Article in English | MEDLINE | ID: mdl-31926186

ABSTRACT

Perfluorooctane sulfonate (PFOS) is a persistent environmental chemical whose biological effects are mediated by multiple mechanisms. Recent evidence suggests that the gut microbiome may be directly impacted by and/or alter the fate and effects of environmental chemicals in the host. Thus, the aim of this study was to determine whether PFOS influences the gut microbiome and its metabolism, and the host metabolome. Four groups of male C57BL/6 J mice were fed a diet with or without 0.003 %, 0.006 %, or 0.012 % PFOS, respectively. 16S rRNA gene sequencing, metabolomic, and molecular analyses were used to examine the gut microbiota of mice after dietary PFOS exposure. Dietary PFOS exposure caused a marked change in the gut microbiome compared to controls. Dietary PFOS also caused dose-dependent changes in hepatic metabolic pathways including those involved in lipid metabolism, oxidative stress, inflammation, TCA cycle, glucose, and amino acid metabolism. Changes in the metabolome correlated with changes in genes that regulate these pathways. Integrative analyses also demonstrated a strong correlation between the alterations in microbiota composition and host metabolic profiles induced by PFOS. Further, using isolated mouse cecal contents, PFOS exposure directly affected the gut microbiota metabolism. Results from these studies demonstrate that the molecular and biochemical changes induced by PFOS are mediated in part by the gut microbiome, which alters gene expression and the host metabolome in mice.


Subject(s)
Alkanesulfonic Acids/toxicity , Fluorocarbons/toxicity , Gastrointestinal Microbiome/drug effects , Animals , Cecum/drug effects , Cecum/metabolism , Cecum/microbiology , Diet , Dose-Response Relationship, Drug , Homeostasis/drug effects , Lipid Metabolism/drug effects , Liver/drug effects , Liver/metabolism , Male , Metabolome , Metabolomics , Mice , Mice, Inbred C57BL , RNA, Ribosomal, 16S/biosynthesis , RNA, Ribosomal, 16S/genetics
14.
Toxicol Sci ; 173(1): 1-2, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31904856
15.
NPJ Precis Oncol ; 3: 26, 2019.
Article in English | MEDLINE | ID: mdl-31602402

ABSTRACT

The peroxisome proliferator-activated-ß/δ (PPARß/δ) was identified in 1994, but not until 1999 was PPARß/δ suggested to be involved in carcinogenesis. Initially, it was hypothesized that expression of PPARß/δ was increased during colon cancer progression, which led to increased transcription of yet-to-be confirmed target genes that promote cell proliferation and tumorigenesis. It was also hypothesized at this time that lipid-metabolizing enzymes generated lipid metabolites that served as ligands for PPARß/δ. These hypothetical mechanisms were attractive because they potentially explained how non-steroidal anti-inflammatory drugs inhibited tumorigenesis by potentially limiting the concentration of endogenous PPARß/δ ligands that could activate this receptor that was increased in cancer cells. However, during the last 20 years, considerable research was undertaken describing expression of PPARß/δ in normal and cancer cells that has led to a significant impact on the mechanisms by which PPARß/δ functions in carcinogenesis. Whereas results from earlier studies led to much uncertainty about the role of PPARß/δ in cancer, more recent analyses of large databases have revealed a more consistent understanding. The focus of this review is on the fundamental level of PPARß/δ expression in normal tissues and cancerous tissue as described by studies during the past two decades and what has been delineated during this timeframe about how PPARß/δ expression influences carcinogenesis, with an emphasis on colon cancer.

16.
Hum Genomics ; 13(1): 27, 2019 06 11.
Article in English | MEDLINE | ID: mdl-31186074

ABSTRACT

The human microbiome is composed of four major areas including intestinal, skin, vaginal, and oral microbiomes, with each area containing unique species and unique functionalities. The human microbiome may be modulated with prebiotics, probiotics, and postbiotics to potentially aid in the treatment of diseases like irritable bowel syndrome, bacterial vaginosis, atopic dermatitis, gingivitis, obesity, or cancer. There is also potential for many of the inhabitants of the human microbiome to directly modulate host gene expression and modulate host detoxifying enzyme activity like cytochrome P450s (CYPs), dehydrogenases, and carboxylesterases. Therefore, the microbiome may be important to consider during drug discovery, risk assessment, and dosing regimens for various diseases given that the human microbiome has been shown to impact host detoxification processes.


Subject(s)
Inactivation, Metabolic/genetics , Microbiota/drug effects , Prebiotics , Probiotics/therapeutic use , Dermatitis, Atopic/drug therapy , Dermatitis, Atopic/genetics , Female , Gingivitis/drug therapy , Gingivitis/genetics , Humans , Irritable Bowel Syndrome/drug therapy , Irritable Bowel Syndrome/genetics , Microbiota/genetics , Vaginosis, Bacterial/drug therapy , Vaginosis, Bacterial/genetics
18.
Mol Carcinog ; 58(9): 1612-1622, 2019 09.
Article in English | MEDLINE | ID: mdl-31062422

ABSTRACT

Considerable progress has been made during the past 20 years towards elucidating the role of peroxisome proliferator-activated receptor-ß/δ (PPARß/δ) in skin cancer. In 1999, the original notion that PPARß/δ was involved with epithelial cell function was postulated based on a correlation between PPARß/δ expression and the induction of messenger RNAs encoding proteins that mediate terminal differentiation in keratinocytes. Subsequent studies definitively revealed that PPARß/δ could induce terminal differentiation and inhibit proliferation of keratinocytes. Molecular mechanisms have since been discovered to explain how this nuclear receptor can be targeted for preventing and treating skin cancer. This includes the regulation of terminal differentiation, mitotic signaling, endoplasmic reticulum stress, and cellular senescence. Interestingly, the effects of activating PPARß/δ can preferentially target keratinocytes with genetic mutations associated with skin cancer. This review provides the history and current understanding of how PPARß/δ can be targeted for both nonmelanoma skin cancer and melanoma and postulates how future approaches that modulate PPARß/δ signaling may be developed for the prevention and treatment of these diseases.


Subject(s)
PPAR delta/metabolism , PPAR-beta/metabolism , Skin Neoplasms/metabolism , Animals , Cell Differentiation/physiology , Cell Proliferation/physiology , Humans , Keratinocytes/metabolism , Melanoma/metabolism , RNA, Messenger/metabolism , Signal Transduction/physiology
19.
Toxicol Sci ; 165(2): 277-283, 2018 10 01.
Article in English | MEDLINE | ID: mdl-30053205

ABSTRACT

An explosion of knowledge on the molecular and cellular mechanisms that mediate carcinogenesis has occurred in recent years. Although cancer has existed for over a million years in the human species, effective cures for most cancers that target molecular and cellular pathways have not been achieved. Multiple cellular targets have been examined for preventing or treating cancers including, but not limited to, transcription factors, kinase-mediated cell signaling pathways, and more recently epigenetic targeting of oncogenes and tumor suppressors, and immunomodulation such as chimeric antigen receptor-T cells. Even as the state of knowledge of cancer mechanisms increases, there is considerable room for improvement in preventing and treating cancers. Understanding how a normal cell is transformed into a cancer cell is known but there is considerable tissue and cell type specificity. This has given rise to the field of precision medicine as applied to cancer therapy. Thus, while the development of preventive and treatment regimens has increased, there are certain obstacles that need to be overcome in order to decrease cancer incidence and increase survival of cancer patients. The purpose of this review is to summarize the advances made in cancer biology and how these advances have been used to develop, and hinder, preventive, and therapeutic strategies for cancer.


Subject(s)
Anticarcinogenic Agents/pharmacology , Carcinogenesis/drug effects , Epigenesis, Genetic/drug effects , Neoplasms/prevention & control , Signal Transduction/drug effects , Carcinogenesis/genetics , Carcinogenesis/metabolism , Cell Transformation, Neoplastic/drug effects , Humans , Molecular Targeted Therapy , Neoplasms/enzymology , Neoplasms/pathology , Xenobiotics/metabolism , Xenobiotics/toxicity
20.
Toxicology ; 404-405: 25-32, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29729928

ABSTRACT

To examine the functional role of peroxisome proliferator-activated receptor-ß/δ (PPARß/δ) and PPARγ in skin cancer, stable cell lines were created in the A431 human squamous cell carcinoma cell line. Expression of PPAR target genes was greatly enhanced in response to ligand activation of PPARß/δ or PPARγ in A431 cells expressing these receptors. PPARß/δ expression blocked the cell cycle at the G2/M phase, and this effect was increased by ligand activation. Ligand activation of PPARß/δ markedly inhibited clonogenicity as compared to vehicle-treated controls. Similarly, ligand activation of PPARγ in A431 cells expressing PPARγ resulted in reduced clonogenicity. Expression of either PPARß/δ or PPARγ markedly reduced tumor volume in ectopic xenografts, while ligand activation of these receptors had little further influence on tumor volume. Collectively, these studies demonstrate that stable expression and activation of PPARß/δ or PPARγ in A431 cells led to reduced tumorigenicity. Importantly, PPAR expression or ligand activation had major impacts on clonogenicity and/or tumor volume. Thus, PPARß/δ or PPARγ could be therapeutically targeted for the treatment of squamous cell carcinomas.


Subject(s)
Carcinogenesis/metabolism , Carcinoma, Squamous Cell/metabolism , Cell Cycle/physiology , PPAR delta/biosynthesis , PPAR-beta/biosynthesis , Skin Neoplasms/metabolism , Animals , Carcinoma, Squamous Cell/prevention & control , Cell Cycle Checkpoints/physiology , Cell Line, Tumor , Female , Humans , Mice, Nude , Skin Neoplasms/prevention & control , Xenograft Model Antitumor Assays/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...