Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 5329 2023-02-24 04:14:55 |
2 format correct Meta information modification 5329 2023-02-24 04:19:54 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Zhao, B.; Xin, Z.; Ren, P.; Wu, H. Subtypes of PPARs and Breast Cancer. Encyclopedia. Available online: https://encyclopedia.pub/entry/41608 (accessed on 03 July 2024).
Zhao B, Xin Z, Ren P, Wu H. Subtypes of PPARs and Breast Cancer. Encyclopedia. Available at: https://encyclopedia.pub/entry/41608. Accessed July 03, 2024.
Zhao, Binggong, Zhiqiang Xin, Ping Ren, Huijian Wu. "Subtypes of PPARs and Breast Cancer" Encyclopedia, https://encyclopedia.pub/entry/41608 (accessed July 03, 2024).
Zhao, B., Xin, Z., Ren, P., & Wu, H. (2023, February 24). Subtypes of PPARs and Breast Cancer. In Encyclopedia. https://encyclopedia.pub/entry/41608
Zhao, Binggong, et al. "Subtypes of PPARs and Breast Cancer." Encyclopedia. Web. 24 February, 2023.
Subtypes of PPARs and Breast Cancer
Edit

Breast cancer is a malignant tumor with high morbidity and lethality. Its pathogenesis is related to the abnormal expression of many genes. The peroxisome proliferator-activated receptors (PPARs) are a class of ligand-dependent transcription factors in the nuclear receptor superfamily. They can regulate the transcription of a large number of target genes, which are involved in life activities such as cell proliferation, differentiation, metabolism, and apoptosis, and regulate physiological processes such as glucose metabolism, lipid metabolism, inflammation, and wound healing. Further, the changes in its expression are associated with various diseases, including breast cancer. The PPARα, PPARβ/δ and PPARγ express differently in different tissues, with differences in target genes, biological activities, and ligand affinities. The PPARs participate in the regulation of carbohydrate and lipid metabolism and homeostasis, as well as various physiological processes such as cell differentiation, proliferation, inflammation, and vascular biology. In addition, the three subtypes of PPARs also regulate the occurrence and development of many malignant tumors via different mechanisms; breast cancer is one of them.

breast cancer PPARs ligands ERs

1. PPARα and Breast Cancer

PPARα, the first PPAR identified, is recognized as an orphan receptor activated by a variety of peroxisome proliferators. The PPARα was originally discovered in rodents and was named for its role in peroxisome proliferation [1]. On the other hand, PPARβ/δ and PPARγ were subsequently discovered and identified as cognate receptors that are activated by distinct peroxisome proliferators [2][3]. However, subsequent research proved that all PPARs fail to play a role in human peroxisome proliferation. PPARα is mainly expressed in metabolically vigorous cells with active fatty acid oxidation capacity, for example in skeletal muscle, brown fat, the liver, heart, and intestinal mucosal tissues [4]. PPARα is of considerable importance to glucose and lipid metabolism and the balance of transport in mammals. Its main function of maintaining lipid homeostasis is realized by increasing cell mobilization, promoting cell uptake, activation, oxidation, and decomposition of fatty acids, and generating ketone bodies for energy production [5]. The ligand-activated PPARα could also catalyze the hydroxylation of fatty acids. Hence, PPARα is the target of fibrates and hypolipidemic drugs for the treatment of abnormal lipid metabolism. The transcription of PPARα is up-regulated by fibrates, which enhance the lipolysis mediated by lipoprotein lipase, promote the oxidative decomposition of fatty acids, and achieve the curative effect of reducing total cholesterol and total triglycerides [6]. Fibrates are effective in increasing insulin sensitivity and protecting the cardiovascular system, so they are also widely used in the clinical treatment of diabetes and cardiovascular diseases [7].
In addition to regulating glucose and lipid metabolism, PPARα plays a role in various cancers. Long-term administration of PPARα agonists was reported as early as 1980 to cause liver cancer in rodents [8]. This effect of agonists was dependent on the receptor PPARα, as they (Wy-14,643 or bezafibrate) did not induce liver cancer in PPARα-null mice [9][10]. The pro-hepatocarcinogenesis effect of PPARα agonists was not evident in humans [11]. The species-specific mechanism of promoting hepatocarcinogenesis is that mouse-derived PPARα rather than human-derived PPARα down-regulated let-7C miRNA to increase the stability of its target gene MYC, an oncogenic factor. The increased expression of MYC promoted hepatocyte mitosis until carcinogenesis [12][13][14]. Some studies have shown increased expression of PPARα in endometrial cancer. Fenofibrate treatment significantly prevented the proliferation of endometrial cancer cells and promoted cell apoptosis [15]. However, other studies have also shown that PPARα knockdown inhibited the proliferation of endometrial cancer cells, promoted cell apoptosis, and reduced the secretion of the angiogenesis-related factor VEGF, while fenofibrate treatment also reduced the secretion of VEGF [16]. Since this contradictory phenomenon is not caused by nonspecificity to PPARα and cytotoxicity at the dose of fenofibrate [17], a possible explanation might be the biphasic response of PPARα activity, i.e., PPARα with very low activity and expression and PPARα with very high activity and expression producing the same effect, known as a U-shaped dose-response curve. PPARα was also aberrantly expressed in melanoma. Fenofibrate treatment inhibited the clone formation and migration abilities of melanoma cells and rendered them highly sensitive to staurosporine (a protein kinase C inhibitor with strong pro-apoptotic activity) [18].
Chang et al. found that, compared to adjacent normal tissues, PPARα and its natural ligand, arachidonic acid (AA), were significantly overexpressed in the tissues of breast cancer. The growth of three breast cancer cells, MDA-MB-231 (ER-), MCF7 (ER++++), and BT-474 (ER++), were stimulated by AA, with the most pronounced pro-proliferative effect on MCF7 cells, revealing a positive correlation between PPARα and the proliferation of ER+ breast cancer cells [19]. Human cytochrome P450 1B1 (CYP1B1)-mediated biotransformation of endogenous estrogens and environmental carcinogens promotes the progression of multiple hormone-dependent tumors, including breast cancer [20]. Hwang et al. found that Wy-14,643 increased CYP1B1 mRNA and protein levels in MCF7 cells and activated PPARα enhanced CYP1B1 promoter activity through directly binding to its PPRE elements [21]. In addition, Castelli et al. found that treatment of breast cancer stem cells with the specific PPARα antagonist GW6471 reduced cell proliferation, viability, and spheroid formation, resulting in metabolic dysfunction and apoptosis [22]. The above experiments in vitro all suggest that PPARα functions in promoting the development of breast cancer. However, Pighetti et al. found that treatment with Wy-14,643 inhibited the ability of DMBA to induce breast tumor formation in rats and induced tumor volume regression [23]. Chandran et al. showed that clofibrate treatment activated the PPARα transcriptional activity and exerted an anti-proliferative effect on breast cancer cells by regulating the levels of tumor suppressors, cell cycle inhibitors, and cell to cycle checkpoint kinases, causing cells to arrest in the G0/G1 phase and significantly inhibiting cell growth. In addition, activated PPARα reduced the expression of inflammatory pathway-related enzymes and their receptors, reduced the protein levels of lipogenic enzymes, regulated the fatty acid oxidation associated gene expression, and affected various lipid metabolism pathways [24]. Yin et al. found that Runt-related transcription factor 2 (RUNX2), with high expression in breast cancer, recruited metastasis-associated 1 (MTA1)/NuRD and the Cullin 4B (CUL4B)-Ring E3 ligase (CRL4B) complex to form a ternary complex. This complex catalyzed histone deacetylation and ubiquitination, inhibited the transcriptional activity of target genes, including PPARα, and promoted the proliferation and invasion of breast cancer cells in vitro. These physiological processes finally led to breast cancer occurrence, bone metastasis, and tumor stemness in vivo (Table 1) [25]. The above findings indicate that PPARα plays a role as a tumor suppressor in breast cancer.
Table 1. The effects of PPARα on breast cancer.
PPARα was generally highly expressed in human primary inflammatory breast cancer cells SUM149PT (3.9-fold higher than primary human breast epithelial cells HMEC) and highly invasive breast cancer cells SUM1315MO2 (3.7-fold higher than HMEC cells) and in human breast tumor tissue (2–6-fold higher than adjacent normal tissues) [24]. The correlation between PPARα and breast cancer is worth further investigation.

2. PPARβ/δ and Breast Cancer

Among the three subtypes of PPARs, PPARβ/δ exhibits higher evolutionary efficiency [1]. In addition, uncoordinated PPARβ/δ also showed more potent transcriptional repression activity. Compared with uncoordinated PPARβ/δ, unligated PPARα and PPARγ do not inhibit PPRE-mediated transcription, which is possibly due to their inability to bind to the nuclear receptor corepressors such as SMRT and NCoR [26]. This relatively rapid rate of evolution and more potent transcriptional repression activity underscore the importance of investigating PPARβ/δ function. The PPARβ/δ are referred to as HUC-1 in humans [27], fatty acid-activated receptors (FAAR) in mice [28], and PPARδ in rats [29]. The PPARβ/δ are widely expressed in most tissues, and their expression level is often higher than that of PPARα and PPARγ. This widespread expression proves its importance in systemic activities and basic cell functions [3][30]. The high baseline expression of PPARγ, especially in the gastrointestinal tract and skeletal muscle, reveals the critical role of PPARβ/δ in fatty acid oxidation and obesity prevention [31]. PPARβ/δ is specific and diversified in cell fate. It can activate housekeeping genes and regulate energy metabolism. In addition, the endogenous natural ligands of PPARβ/δ are very broad and non-specific. The ability of these ligands to activate PPARβ/δ is relatively weak. Therefore, the physiological function of PPARβ/δ is difficult to simplify. Without ligand binding, PPARβ/δ degrades fast, while ligand binding inhibits ubiquitin-proteasome activity, thereby extending its half-life [32][33]. This phenomenon may also be attributed to ligand-induced PPARβ/δ expression [34]. Ligand-activated PPARβ/δ could increase the levels of serum high-density lipoprotein cholesterol, decrease the levels of serum triglycerides in mice [35], non-human primates [36], and humans [37], and improve the metabolic syndrome such as obesity and insulin resistance induced by a high-fat diet or genetic predisposition [31][38]. Inhibition of insulin resistance by activated PPARβ/δ might also improve progressive neurodegeneration and its associated learning and memory deficits and prevent Alzheimer’s disease [39][40]. In addition, PPARβ/δ also have considerable preventive or therapeutic capacity against genetic [41], diet [42], or chemically induced [43] liver inflammation.
The above evidence supports the development of PPARβ/δ specific agonists acting as clinical drugs for the treatment of diseases such as obesity, diabetes, metabolic syndrome, and liver inflammation. However, the synthesis of PPARβ/δ-targeted drugs has encountered significant obstacles related to clinical safety due to substantial controversy regarding the reports on the role of PPARβ/δ in cancer [44][45]. Ligand-activated PPARβ/δ could promote terminal differentiation of keratinocytes [46], enhance lipid deposition [47], inhibit cell proliferation [48], and inhibit the progression of skin cancers such as psoriasis. However, it has also been shown that transgenic mice that induced activation of PPARβ/δ in the epidermis developed an inflammatory skin disease strikingly similar to psoriasis. These mice were characterized by hyperproliferation of keratinocytes, aggregation of dendritic cells, and endothelial cell activation. The gene dysregulation and activation of key transcriptional programs and Th17 subsets of T cells in transgenic mice were also highly similar to psoriasis [49]. In addition, PPARβ/δ activated by UV stimulation directly promoted the expression of oncogene Src and upregulated its kinase activity, enhanced the EGFR/ERK1/2 signaling pathway, and promoted epithelial-mesenchymal transition (EMT), which promotes keratinocyte differentiation and proliferation [50]. This result also reveals the cancer-promoting effect of PPARβ/δ on skin cancer. A possible and one-sided explanation for this contradiction was that activation of PPARβ/δ existed both in keratinocytes and adjacent fibroblasts. The PPARβ/δ in fibroblasts inhibited IL-1 signaling by directly upregulating the expression of secreted interleukin-1 receptor antagonist (sIL-1ra), thereby regulating keratinocyte proliferation [51]. In addition to skin cancer, the PPARβ/δ also have a controversial role in colorectal cancer [52][53][54].
Human genome PPARβ/δ is located at 6p21.2, an increased site for ER- and high-risk breast cancer [55], which reveals the correlation between PPARβ/δ and breast cancer. PPARβ/δ was highly expressed in the nucleus in human normal breast epithelial cells and weakly expressed or even absent in 92% of human breast lobular and ductal cancer cells [56][57][58]. The expression of PPARβ/δ in mouse malignant breast cancer cells C20 was also significantly lower than that in mouse keratinocytes (nearly 4-fold) and human normal mammary epithelial cells MCF10A (more than 2-fold) [59]. The patients’ survival rate with breast cancer and the expression of PPARβ/δ have a negative correlation [60]. In 2004, Stephen et al. reported for the first time that PPARβ/δ activated by specific ligand compound F or GW501,516 could promote the proliferation of ER+ breast cancer cells MCF7 and T47D. It could also promote in T47D cells vascular endothelial growth factor α (VEGFα) and its receptor FLT-1 and encourage the proliferation of human umbilical vein endothelial cells (HUVEG) in vitro. However, activated PPARβ/δ did not exert similar effects on ER- breast cancer cells MDA-MB-231 and BT-20, revealing that the pro-proliferative and pro-angiogenic effects of PPARβ/δ on breast cancer are dependent on ER [61]. Conversely, in 2008, Girroir et al. reported that PPARβ/δ was activated by specific ligands (GW0742 or GW501,516) and inhibited the growth of MCF7 cells [62]. In 2010, Foreman et al. reported that PPARβ/δ activated by the above two ligands also inhibited proliferation and clone formation and promoted apoptosis in mouse C20 cells [59]. Additionally, in 2014, Yao et al. reported that the overexpression of PPARβ/δ prevented the proliferation of breast cancer cells, MDA-MB-231 and MCF7, while the treatment of the agonist GW0742 further inhibited the proliferation of MCF7 cells without any effect on the MDA-MB-231 cells. The overexpression of PPARβ/δ inhibited the clone formation of these two cell lines, while further treatment with GW0742 inhibited the clone formation of MDA-MB-231 cells significantly more than that of MCF7 cells. However, the overexpression or ligand-activated of PPARβ/δ did not affect apoptosis in either of the two breast cancer cell lines. Further, the overexpression of PPARβ/δ could inhibit the growth of xenograft tumor in MDA-MB-231 cells better than in MCF7 cells, and treatment with GW0742 further inhibited the volume of mouse xenografts [63]. These findings, although inconsistent with Stephen’s report [61], also confirm that the effects of PPARβ/δ on ER+ and ER- breast cancer cells were different. However, by real-time analysis of cell doubling time, Palkar et al. found that neither GW0742-activated nor highly specific irreversible antagonist GSK3787 inhibited PPARβ/δ had effects on the proliferation of MCF7 cells, despite the fact that both of them had the converse effect on the mRNA level of PPARβ/δ target gene Angptl4 in vitro and in vivo [64]. Additionally, although these disparate results may be attributed to the concentration of ligands used, cell treatment time, cell proliferation assessment methods, etc., the exact function of PPARβ/δ on breast cancer cell apoptosis and proliferation remains unclarified so far. Several experiments are required to reach consensus.
Retinoic acid (RA) as a tumor suppressor exhibits potent anticancer activity mediated by the nuclear retinoic acid receptor (RAR). The intracellular lipid-binding protein cellular retinoic acid-binding protein II (CRABP-II) targets RA to the RAR, while another lipid-binding protein, fatty acid binding protein 5 (FABP5), could deliver it to the non-canonical RA receptor PPARβ /δ. The FABP5/CRABP-II ratio determines the partition of RA between the two receptors. Noy’s team constructed two breast cancer MMTV-neu transgenic mouse models expressing different FABP5/CRABP-II ratios in breast tissue. It was observed that transgenic mice with a high FABP5/CRABP-II ratio produced larger breast tumors. On the contrary, the reduction of this ratio resulted in the suppression of breast tumor growth and gene expression, including PDK1 and cell proliferation-related genes, through the transfer of RA signaling from PPARβ/δ to RAR. This study proposes a new mechanism by which PPARβ/δ promote breast cancer [65]. Additionally, the epidermal growth factor receptor (EGFR) as a tumor-promoting factor can promote breast cancer cell proliferation and induce breast tumorigenesis. Noy’s team also found that treatment of MCF7 cells with the EGFR ligand heregulin-β1 could directly upregulate the expression of FABP5 and PDK1. The results indicated that FABP5 and PPARβ/δ were the key mediators of EGFR’s ability to enhance cell proliferation, further confirming that PPARβ/δ acted as a tumor-promoting factor playing a role in breast cancer [66]. However, studies on human keratinocyte HaCaT found that FABP5 neither delivered RA to PPARβ/δ nor promoted anti-apoptotic activity by upregulating PDK1 levels. This phenomenon was also identified in HaCaT cells that stably overexpress PPARβ/δ [67]. The above results suggest that the cancer-promoting effect of RA-mediated PPARβ/δ may be specific to breast cancer [68]. Wang et al. found that PPARβ/δ could promote the survival of MCF7 cells under rough microenvironmental conditions by reducing oxidative stress and promoting AKT-mediated survival signaling [69]. The correlation between PPARβ/δ and PDK1 is currently controversial. Although the above studies have found that the expression levels of the two are correlated, there are also studies showing that PDK1 is not a target gene of PPARβ/δ [54][68][70]. In addition to the research around the effect of PPARβ/δ on the proliferation and apoptosis of breast cancer cells, scholars have found that PPARβ/δ also has an effect on the invasion and metastasis of breast cancer cells. Adhikary found that PPARβ/δ, specifically antagonized by ST247 and DG172, inhibited serum and transforming growth factor β (TGFβ)-induced invasion of MDA-MB-231 cells [71]. However, Wang uncovered that the PPARβ/δ expression levels in more metastatic breast cancer basal cell lines were significantly higher than those in luminal cells. Additionally, after the inoculation with MCF7 cells overexpressing PPARβ/δ, the breast tumor volume and lung metastasis of mice increased significantly (Table 2) [69]. In conclusion, the exact role of PPARβ/δ on breast cancer still requires more experimental studies.
Table 2. The effects of PPARβ/δ on breast cancer.

3. PPARγ and Breast Cancer

PPARγ1 and PPARγ2 are two isoforms of PPARγ, that were found in mice. The PPARγ2 mRNA was the predominant PPAR isoform in mouse mammary tissues [76]. In humans and monkeys, in addition to PPARγ1 and PPARγ2, a third isoform of PPARγ4 was found. These isoforms are the transcripts of seven mRNA spliceosomes (PPARγ1, PPARγ2, PPARγ3, PPARγ4, PPARγ5, PPARγ6, and PPARγ7) from the different transcription start sites, which are transcribed through alternative splicing of exons in the 5’-terminal region (A1, A2, B, C, and D) [77]. The PPARγ1, PPARγ3, PPARγ5, and PPARγ7 mRNAs translate into the same protein, PPARγ1, while PPARγ2 mRNA translates into PPARγ2 protein, whereas PPARγ4 and PPARγ6 mRNAs translate into the same PPARγ4 protein. PPARγ1 is expressed in almost all tissues, with the highest level in white and brown adipose tissues. Under normal physiological conditions, the larger PPARγ2 isoform (with additional amino acids at the amino-terminal of PPARγ2, 30 in mice and 28 in humans) is only expressed in brown and white adipose tissue, whereas its expression in the liver and skeletal muscle is caused by excessive caloric intake or genetic obesity. PPARγ4 is under-researched and expressed in macrophages and adipose tissues [78][79][80]. PPARγ widely expressed in white and brown adipose tissues, the large intestine, and the spleen. However, PPARγ is also found in the liver, pancreas, and tissues of the immune system [81]. A considerable number of studies have confirmed that ligand-activated PPARγ could regulate fat distribution and glucose and lipid metabolism [82] and reduce the inflammatory response of cardiovascular cells, especially endothelial cells [83]. Its specific agonist is relatively effective in the treatment of hyperlipidemia, hyperglycemia, and cardiovascular disease [84]. The specific agonists of PPARγ, i.e., TZDs, are clinical drugs currently on the market as insulin sensitizers for the treatment of type 2 diabetes, targeting PPARγ to exert a hypoglycemic effect. The antidiabetic activity of TZDs was first discovered in the early 1980s [85][86][87][88]. PPARγ is also involved in neural differentiation during the formation of neural precursor cells [89]. Therefore, its specific agonists could also act as protective agents for neurons, inducing synaptic plasticity and neurite outgrowth, and improving the symptoms of some neurological diseases [90]. In addition to the above effects, a large number of reports also pointed out that ligand-activated PPARγ exerts anti-tumor effects by promoting cell apoptosis and preventing cell proliferation, regulating cell metastasis, and stimulating angiogenesis, thereby inhibiting the occurrence and development of tumors of the liver [91], bladder [92], lung [93][94], brain [95], thyroid [96], esophagus [97] and colorectum [98][99][100][101].
PPARγ also plays a role in breast cancer progression. In 1998, it was reported that TZD-activated PPARγ could induce terminal differentiation of malignant mammary epithelial cells in vitro [102]. However, in 1999, researchers found that ligand-activated PPARγ could prevent the development of experimental breast cancer in vivo. The report showed that GW7845 as an activator of PPARγ significantly inhibited nitrosomethylurea (NMU)-induced mammary tumor incidence, tumor number, and tumor weight in rats [103]. Subsequent reports of ligand-activated PPARγ inhibiting breast cancer development have experienced a rise. A 2001 study showed that TGZ inhibited DMBA-induced mammary tumor progression in rats, reduced malignancy incidence, and induced regression or stasis of total tumor volume [23]. A study in 2009 showed that the conjugated fatty acid α-eleostearic acid (α-ESA) could act as an agonist of PPARγ, upregulating the level of PPARγ mRNA in MCF7 cells, upregulating PPARγ’s DNA binding activity and transcriptional activity, and mediating PPARγ nuclear translocation, thereby reducing MCF7 cell viability and promoting tumor cell apoptosis. At the same time, α-ESA-induced high PPARγ expression was associated with an inhibitory effect on ERK1/2 MAPK phosphorylation activation. This suggests that pERK1/2 might play a negative regulatory role on PPARγ levels [104]. Bonofiglio’s team discovered an important pathway for PPARγ in human breast cancer cell growth, cycle arrest, and apoptosis. RGZ-activated PPARγ inhibits the PI3K/AKT pathway and induces the antiproliferative effect of MCF7 cells [105]. RGZ also increased the binding of PPARγ to the NF-κB sequence on the promoter sequence of p53, upregulated the expression level of p53 in MCF7, induced caspase 9 cleavage and DNA fragmentation, triggered the apoptotic pathway, stopped the growth, and promoted apoptosis of breast cancer cells [106]. Furthermore, in several breast cancer cell lines, RGZ activated the human Fas ligand (FasL) promoter in a PPARγ-dependent manner, increased the binding of PPARγ with Sp1 to the Sp1 sequence located within the FasL promoter, and positively regulated FasL expression [107]. FasL is a type II transmembrane protein expressed on the membrane surface of activated T lymphocytes and cancer cells. By binding to its receptor Fas [108][109], it activates the cascade of caspases and induces apoptosis [110]. These studies reveal a novel molecular mechanism by which PPARγ induces growth arrest and apoptosis in breast cancer cells. An in vivo study in 2011 showed that TZD-activated PPARγ inhibited MAPK/STAT3/AKT phosphorylation-mediated leptin signaling in MCF7 cells. On one hand, this effect led to the inhibition of MCF7 xenografts through the counteraction of the stimulatory effects of leptin on estrogen signaling. On the other hand, it inhibited leptin-induced cell-cell aggregation and tumor cell proliferation, exerting pro-apoptotic and anti-proliferative effects on breast cancer cell lines [111].
In addition to its ligand-activated state, PPARγ also involves itself in the development of breast cancer in a non-ligand-independent manner. The PPARs and ERα are both members of the nuclear receptor superfamily. The ERα signaling pathway has a critical role in metabolism regulation and various physiological processes in the development of breast cancer [112][113]. Bonofiglio’s team found for the first time that ERα could bind to the PPRE element to inhibit its mediated transcriptional activity independently of PPARs. Interestingly, PPAR/RXR heterodimers could also bind to the ER response element (ERE) independently of ERs [114]. PPARγ physically interacted with ERα to form a ternary complex with a regulatory subunit of PI3K and p85. PPARγ and ERα played opposite roles in the regulation of PI3K/AKT signaling, which involves cell survival and proliferation [105]. The crosstalk between the PPARγ and ERα signaling pathways revealed the important role of PPARγ in the development of ER+ breast cancer. Since PPARγ-null mice are embryonic lethal, scientists have developed other ways to create transgenic animal models that silence PPARγ. Yin et al. investigated the susceptibility of PPARγ inactivation to MPA- and DMBA-induced breast cancer in mice by constructing an MMTV-Pax8PPARγ transgenic mouse model. In the absence of induction, the mammary glands of transgenic and wild-type mice did not differ in functional development or propensity for tumor formation, a finding consistent with Cui et al.’s [115]. However, after being induced by MPA and DMBA, transgenic mice developed higher tumor diversity than wild-type mice. These tumors were predominantly ER+ ductal breast cancers, further revealing the role of PPARγ in the development of ER+ breast cancer. The decrease in PTEN expression, the induction of pERK1 and pAKT levels, and decreasing pGSK3β level, Pax8PPARγ promotes Wnt signaling [116]. However, in constructing transgenic mice with constitutively active forms of MMTV-VpPPARγ, Saez et al. found that activation of PPARγ signaling did not affect mammary gland development in transgenic mice, which had no phenotypic difference with wild-type mice. On the other hand, when such transgenic mice were crossed with breast cancer-prone transgenic MMTV-PyV mice, the progeny biogenic mice developed tumors much faster and with a higher degree of malignancy and differentiation of the tumors. This molecular mechanism for promoting breast cancer development might also be attributed to the promotion of PPARγ on the Wnt signaling pathway [117]. Tian et al. conducted a parallel experiment on immunocompetent FVB mice, with one group of implanted tumor cells transduced with wild-type PPARγ, and the other with constitutively active PPARγCA. They found that the growth of mammary tumors in mice implanted with PPARγCA-transduced cells was enhanced, which was correlated with endothelial stem cells and angiogenesis increasing. PPARγCA induced ErbB2-transformed mammary epithelial cells to secrete Angptl4 protein, which enhanced angiogenesis in vivo and promoted tumor growth [118]. The above studies based on animal models reveal the contradictory roles (either inhibiting or promoting) of PPARγ in the occurrence and development of breast cancer. The potential reasons for this discrepancy remain to be investigated. The possible causes could be traced to the differences in the construction of animal models or the difference in the length of experimental periods. In addition, a 2019 study showed that PPARγ directly bound to the PPRE element of the protein tyrosine phosphatase receptor-type F (PTPRF) promoter and recruited RNA polymerase II and H3K4me3 to promote the transcription of PTPRF. These processes inhibited breast cancer cell proliferation and migration in vitro and inhibited breast tumor growth and distant metastasis in mice [119]. A 2020 experiment in vitro showed that PPARγ, which is commonly expressed in human primary and metastatic breast cancer [120], interacted with Nur77, recruited the ubiquitin E3 enzyme Trim13 to target the ubiquitin proteasomal degradation of Nur77, and promoted breast cancer progression. Nur77, a tumor suppressor, inhibits breast cancer cells from uptaking exogenous fatty acids and blocks the accumulation of fatty acids in the tumor metabolic microenvironment by inhibiting the transcription of the transmembrane protein CD36 and the cytoplasmic fatty acid-binding protein FABP4. Therefore, blocking the interaction between PPARγ and Nur77 can be used as a clinical approach for PPARγ ligand-independent treatment of breast cancer (Table 3) [121]. However, due to the relatively high concentrations of endogenous natural ligands in cells, it remains to be verified whether these conclusions are truly ligand-independent of PPARγ.
Table 3. The effects of PPARγ on breast cancer.
In 2005, an immunohistochemical test of 170 patients with invasive breast cancer showed that the expression of PPARγ was negatively associated with histological grade (p = 0.019). PPARγ had a significantly favorable effect on recurrence-free survival in breast ductal carcinoma patients (p = 0.027) and was an independent prognostic factor in ductal carcinoma patients (p = 0.039) [122]. In 2008, a study presented that the nuclear expression of PPARγ had a preventive effect on the recurrence of female breast ductal carcinoma in situ. Its expression level was negatively correlated with tumor recurrence (p = 0.024) [123]. These clinical research studies and the above experimental results reveal the important function of PPARγ in the occurrence and development of breast cancer. The overexpression of PPARγ in breast tumors and the physiological effects of its ligands on breast cancer cells indicate that PPARγ will be a possible target in breast cancer clinical prevention and treatment.

4. PPARs and TNBC

TNBC, the most aggressive subtype of breast cancer, has no effect on hormone therapy or HER2-targeted therapy due to its lack of the three receptors. Surgery or chemotherapy, the only viable option, is a systemic therapy that causes not only physical distress but a poor prognosis for TNBC patients [124]. Therefore, it is very necessary to explore new treatment methods or target drugs to improve the prognosis of TNBC. Li et al. found that the PPARα-specific agonist fenofibrate had anti-proliferative effects on breast cancer cell lines, and the top 5 most sensitive cells are all TNBC cell lines [125]. Kwong found that fatty acid binding protein 7 (FABP7) failed to induce the efficient use of glucose to generate ATP in the TNBC cell line Hs578T during serum starvation, eventually leading to cell death. This metabolic effect of FABP7 on Hs578T cells was mediated by PPARα [126]. Studies by Stephen’s group showed that PPARβ/δ activated by GW501,516 could promote the proliferation of MCF7 and T47D cells, but it had no similar effect on the TNBC cell lines MDA-MB-231 and BT-20 [61]. The expression level of PPARβ/δ in highly aggressive basal cells was significantly higher than that in luminal cells [69]. In addition, Adhikary’s team found that ST247 and DG172 specifically antagonized PPARβ/δ strongly inhibited the invasion ability of MDA-MB-231 cells induced by serum and TGFβ [71]. Jiang’s team found that the expression of PPARγ in the breast tissues of TNBC patients was significantly lower than that of other subtype patients, and its expression in MDA-MB-231 cells was also significantly lower than that of other breast cancer cell lines. Previous studies have reported that the PPARγ-specific agonist RGZ had antitumor effects in breast cancer. However, it did not exert significant anti-proliferative effects on MDA-MB-231 cells. RGZ combined with the demethylation agent hydralazine significantly inhibited the proliferation of MDA-MB-231 cells and promoted cell apoptosis [127]. Apaya et al. showed that epoxy-eicosatrienoic acid (EET) induced the nuclear translocation of FABP4 and FABP5 in MDA-MB-231 cells, thereby promoting the nuclear accumulation of PPARγ and affecting cell proliferation and migration [128]. These results reveal the important roles of all three subtypes of PPARs and their ligands in TNBC and suggest that more attention should be directed to drug combination therapies against TNBC.

References

  1. Issemann, I.; Green, S. Activation of a member of the steroid hormone receptor superfamily by peroxisome proliferators. Nature 1990, 347, 645–650.
  2. Dreyer, C.; Krey, G.; Keller, H.; Givel, F.; Helftenbein, G.; Wahli, W. Control of the peroxisomal beta-oxidation pathway by a novel family of nuclear hormone receptors. Cell 1992, 68, 879–887.
  3. Kliewer, S.A.; Forman, B.M.; Blumberg, B.; Ong, E.S.; Borgmeyer, U.; Mangelsdorf, D.J.; Umesono, K.; Evans, R.M. Differential expression and activation of a family of murine peroxisome proliferator-activated receptors. Proc. Natl. Acad. Sci. USA 1994, 91, 7355–7359.
  4. Escher, P.; Braissant, O.; Basu-Modak, S.; Michalik, L.; Wahli, W.; Desvergne, B. Rat PPARs: Quantitative analysis in adult rat tissues and regulation in fasting and refeeding. Endocrinology 2001, 142, 4195–4202.
  5. Pyper, S.R.; Viswakarma, N.; Yu, S.; Reddy, J.K. PPARalpha: Energy combustion, hypolipidemia, inflammation and cancer. Nucl. Recept. Signal. 2010, 8, e002.
  6. Staels, B.; Dallongeville, J.; Auwerx, J.; Schoonjans, K.; Leitersdorf, E.; Fruchart, J.C. Mechanism of action of fibrates on lipid and lipoprotein metabolism. Circulation 1998, 98, 2088–2093.
  7. Balfour, J.A.; McTavish, D.; Heel, R.C. Fenofibrate. A review of its pharmacodynamic and pharmacokinetic properties and therapeutic use in dyslipidaemia. Drugs 1990, 40, 260–290.
  8. Reddy, J.K.; Azarnoff, D.L.; Hignite, C.E. Hypolipidaemic hepatic peroxisome proliferators form a novel class of chemical carcinogens. Nature 1980, 283, 397–398.
  9. Peters, J.M.; Cattley, R.C.; Gonzalez, F.J. Role of PPAR alpha in the mechanism of action of the nongenotoxic carcinogen and peroxisome proliferator Wy-14,643. Carcinogenesis 1997, 18, 2029–2033.
  10. Hays, T.; Rusyn, I.; Burns, A.M.; Kennett, M.J.; Ward, J.M.; Gonzalez, F.J.; Peters, J.M. Role of peroxisome proliferator-activated receptor-alpha (PPARalpha) in bezafibrate-induced hepatocarcinogenesis and cholestasis. Carcinogenesis 2005, 26, 219–227.
  11. Peters, J.M.; Cheung, C.; Gonzalez, F.J. Peroxisome proliferator-activated receptor-alpha and liver cancer: Where do we stand? J. Mol. Med. 2005, 83, 774–785.
  12. Shah, Y.M.; Morimura, K.; Yang, Q.; Tanabe, T.; Takagi, M.; Gonzalez, F.J. Peroxisome proliferator-activated receptor alpha regulates a microRNA-mediated signaling cascade responsible for hepatocellular proliferation. Mol. Cell. Biol. 2007, 27, 4238–4247.
  13. Cheung, C.; Akiyama, T.E.; Ward, J.M.; Nicol, C.J.; Feigenbaum, L.; Vinson, C.; Gonzalez, F.J. Diminished hepatocellular proliferation in mice humanized for the nuclear receptor peroxisome proliferator-activated receptor alpha. Cancer Res. 2004, 64, 3849–3854.
  14. Morimura, K.; Cheung, C.; Ward, J.M.; Reddy, J.K.; Gonzalez, F.J. Differential susceptibility of mice humanized for peroxisome proliferator-activated receptor alpha to Wy-14,643-induced liver tumorigenesis. Carcinogenesis 2006, 27, 1074–1080.
  15. Holland, C.M.; Saidi, S.A.; Evans, A.L.; Sharkey, A.M.; Latimer, J.A.; Crawford, R.A.F.; Charnock-Jones, D.S.; Print, C.G.; Smith, S.K. Transcriptome analysis of endometrial cancer identifies peroxisome proliferator-activated receptors as potential therapeutic targets. Mol. Cancer Ther. 2004, 3, 993–1001.
  16. Nickkho-Amiry, M.; McVey, R.; Holland, C. Peroxisome proliferator-activated receptors modulate proliferation and angiogenesis in human endometrial carcinoma. Mol. Cancer Res. 2012, 10, 441–453.
  17. Jiao, H.; Zhao, B. Cytotoxic effect of peroxisome proliferator fenofibrate on human HepG2 hepatoma cell line and relevant mechanisms. Toxicol. Appl. Pharmacol. 2002, 185, 172–179.
  18. Grabacka, M.; Plonka, P.M.; Urbanska, K.; Reiss, K. Peroxisome proliferator-activated receptor alpha activation decreases metastatic potential of melanoma cells in vitro via down-regulation of Akt. Clin. Cancer Res. 2006, 12, 3028–3036.
  19. Chang, N.-W.; Wu, C.-T.; Chen, D.-R.; Yeh, C.-Y.; Lin, C. High levels of arachidonic acid and peroxisome proliferator-activated receptor-alpha in breast cancer tissues are associated with promoting cancer cell proliferation. J. Nutr. Biochem. 2013, 24, 274–281.
  20. Cui, J.; Meng, Q.; Zhang, X.; Cui, Q.; Zhou, W.; Li, S. Design and Synthesis of New α-Naphthoflavones as Cytochrome P450 (CYP) 1B1 Inhibitors To Overcome Docetaxel-Resistance Associated with CYP1B1 Overexpression. J. Med. Chem. 2015, 58, 3534–3547.
  21. Hwang, Y.P.; Won, S.S.; Jin, S.W.; Lee, G.H.; Pham, T.H.; Choi, J.H.; Kang, K.W.; Jeong, H.G. WY-14643 Regulates CYP1B1 Expression through Peroxisome Proliferator-Activated Receptor α-Mediated Signaling in Human Breast Cancer Cells. Int. J. Mol. Sci. 2019, 20, 5928.
  22. Castelli, V.; Catanesi, M.; Alfonsetti, M.; Laezza, C.; Lombardi, F.; Cinque, B.; Cifone, M.G.; Ippoliti, R.; Benedetti, E.; Cimini, A.; et al. PPARα-Selective Antagonist GW6471 Inhibits Cell Growth in Breast Cancer Stem Cells Inducing Energy Imbalance and Metabolic Stress. Biomedicines 2021, 9, 127.
  23. Pighetti, G.M.; Novosad, W.; Nicholson, C.; Hitt, D.C.; Hansens, C.; Hollingsworth, A.B.; Lerner, M.L.; Brackett, D.; Lightfoot, S.A.; Gimble, J.M. Therapeutic treatment of DMBA-induced mammary tumors with PPAR ligands. Anticancer Res. 2001, 21, 825–829.
  24. Chandran, K.; Goswami, S.; Sharma-Walia, N. Implications of a peroxisome proliferator-activated receptor alpha (PPARα) ligand clofibrate in breast cancer. Oncotarget 2016, 7, 15577–15599.
  25. Yin, X.; Teng, X.; Ma, T.; Yang, T.; Zhang, J.; Huo, M.; Liu, W.; Yang, Y.; Yuan, B.; Yu, H.; et al. RUNX2 recruits the NuRD(MTA1)/CRL4B complex to promote breast cancer progression and bone metastasis. Cell Death Differ. 2022, 29, 2203–2217.
  26. Shi, Y.; Hon, M.; Evans, R.M. The peroxisome proliferator-activated receptor delta, an integrator of transcriptional repression and nuclear receptor signaling. Proc. Natl. Acad. Sci. USA 2002, 99, 2613–2618.
  27. Schmidt, A.; Endo, N.; Rutledge, S.J.; Vogel, R.; Shinar, D.; Rodan, G.A. Identification of a new member of the steroid hormone receptor superfamily that is activated by a peroxisome proliferator and fatty acids. Mol. Endocrinol. 1992, 6, 1634–1641.
  28. Amri, E.Z.; Bonino, F.; Ailhaud, G.; Abumrad, N.A.; Grimaldi, P.A. Cloning of a protein that mediates transcriptional effects of fatty acids in preadipocytes. Homology to peroxisome proliferator-activated receptors. J. Biol. Chem. 1995, 270, 2367–2371.
  29. Evans, R.M.; Barish, G.D.; Wang, Y.-X. PPARs and the complex journey to obesity. Nat. Med. 2004, 10, 355–361.
  30. Braissant, O.; Foufelle, F.; Scotto, C.; Dauça, M.; Wahli, W. Differential expression of peroxisome proliferator-activated receptors (PPARs): Tissue distribution of PPAR-alpha, -beta, and -gamma in the adult rat. Endocrinology 1996, 137, 354–366.
  31. Wang, Y.-X.; Lee, C.-H.; Tiep, S.; Yu, R.T.; Ham, J.; Kang, H.; Evans, R.M. Peroxisome-proliferator-activated receptor delta activates fat metabolism to prevent obesity. Cell 2003, 113, 159–170.
  32. Genini, D.; Catapano, C.V. Block of nuclear receptor ubiquitination. A mechanism of ligand-dependent control of peroxisome proliferator-activated receptor delta activity. J. Biol. Chem. 2007, 282, 11776–11785.
  33. Wadosky, K.M.; Willis, M.S. The story so far: Post-translational regulation of peroxisome proliferator-activated receptors by ubiquitination and SUMOylation. Am. J. Physiol. Heart Circ. Physiol. 2012, 302, H515–H526.
  34. Rieck, M.; Wedeken, L.; Müller-Brüsselbach, S.; Meissner, W.; Müller, R. Expression level and agonist-binding affect the turnover, ubiquitination and complex formation of peroxisome proliferator activated receptor beta. FEBS J. 2007, 274, 5068–5076.
  35. Leibowitz, M.D.; Fiévet, C.; Hennuyer, N.; Peinado-Onsurbe, J.; Duez, H.; Bergera, J.; Cullinan, C.A.; Sparrow, C.P.; Baffic, J.; Berger, G.D.; et al. Activation of PPARdelta alters lipid metabolism in db/db mice. FEBS Lett. 2000, 473, 333–336.
  36. Oliver, W.R.J.; Shenk, J.L.; Snaith, M.R.; Russell, C.S.; Plunket, K.D.; Bodkin, N.L.; Lewis, M.C.; Winegar, D.A.; Sznaidman, M.L.; Lambert, M.H.; et al. A selective peroxisome proliferator-activated receptor delta agonist promotes reverse cholesterol transport. Proc. Natl. Acad. Sci. USA 2001, 98, 5306–5311.
  37. Sprecher, D.L.; Massien, C.; Pearce, G.; Billin, A.N.; Perlstein, I.; Willson, T.M.; Hassall, D.G.; Ancellin, N.; Patterson, S.D.; Lobe, D.C.; et al. Triglyceride:high-density lipoprotein cholesterol effects in healthy subjects administered a peroxisome proliferator activated receptor delta agonist. Arterioscler. Thromb. Vasc. Biol. 2007, 27, 359–365.
  38. Tanaka, T.; Yamamoto, J.; Iwasaki, S.; Asaba, H.; Hamura, H.; Ikeda, Y.; Watanabe, M.; Magoori, K.; Ioka, R.X.; Tachibana, K.; et al. Activation of peroxisome proliferator-activated receptor delta induces fatty acid beta-oxidation in skeletal muscle and attenuates metabolic syndrome. Proc. Natl. Acad. Sci. USA 2003, 100, 15924–15929.
  39. de la Monte, S.M.; Wands, J.R. Alzheimer’s disease is type 3 diabetes-evidence reviewed. J. Diabetes Sci. Technol. 2008, 2, 1101–1113.
  40. de la Monte, S.M.; Tong, M.; Lester-Coll, N.; Plater, M.J.; Wands, J.R. Therapeutic rescue of neurodegeneration in experimental type 3 diabetes: Relevance to Alzheimer’s disease. J. Alzheimers Dis. 2006, 10, 89–109.
  41. Gross, B.; Hennuyer, N.; Bouchaert, E.; Rommens, C.; Grillot, D.; Mezdour, H.; Staels, B. Generation and characterization of a humanized PPARδ mouse model. Br. J. Pharmacol. 2011, 164, 192–208.
  42. Liu, S.; Hatano, B.; Zhao, M.; Yen, C.-C.; Kang, K.; Reilly, S.M.; Gangl, M.R.; Gorgun, C.; Balschi, J.A.; Ntambi, J.M.; et al. Role of peroxisome proliferator-activated receptor / in hepatic metabolic regulation. J. Biol. Chem. 2011, 286, 1237–1247.
  43. Shan, W.; Nicol, C.J.; Ito, S.; Bility, M.T.; Kennett, M.J.; Ward, J.M.; Gonzalez, F.J.; Peters, J.M. Peroxisome proliferator-activated receptor-beta/delta protects against chemically induced liver toxicity in mice. Hepatology 2008, 47, 225–235.
  44. Peters, J.M.; Gonzalez, F.J. Sorting out the functional role(s) of peroxisome proliferator-activated receptor-beta/delta (PPARbeta/delta) in cell proliferation and cancer. Biochim. Biophys. Acta 2009, 1796, 230–241.
  45. Peters, J.M.; Hollingshead, H.E.; Gonzalez, F.J. Role of peroxisome-proliferator-activated receptor beta/delta (PPARbeta/delta) in gastrointestinal tract function and disease. Clin. Sci. 2008, 115, 107–127.
  46. Tan, N.S.; Michalik, L.; Noy, N.; Yasmin, R.; Pacot, C.; Heim, M.; Flühmann, B.; Desvergne, B.; Wahli, W. Critical roles of PPAR beta/delta in keratinocyte response to inflammation. Genes Dev. 2001, 15, 3263–3277.
  47. Schmuth, M.; Haqq, C.M.; Cairns, W.J.; Holder, J.C.; Dorsam, S.; Chang, S.; Lau, P.; Fowler, A.J.; Chuang, G.; Moser, A.H.; et al. Peroxisome proliferator-activated receptor (PPAR)-beta/delta stimulates differentiation and lipid accumulation in keratinocytes. J. Investig. Dermatol. 2004, 122, 971–983.
  48. Kim, D.J.; Bility, M.T.; Billin, A.N.; Willson, T.M.; Gonzalez, F.J.; Peters, J.M. PPARbeta/delta selectively induces differentiation and inhibits cell proliferation. Cell Death Differ. 2006, 13, 53–60.
  49. Romanowska, M.; Reilly, L.; Palmer, C.N.A.; Gustafsson, M.C.U.; Foerster, J. Activation of PPARbeta/delta causes a psoriasis-like skin disease in vivo. PLoS ONE 2010, 5, e9701.
  50. Montagner, A.; Delgado, M.B.; Tallichet-Blanc, C.; Chan, J.S.K.; Sng, M.K.; Mottaz, H.; Degueurce, G.; Lippi, Y.; Moret, C.; Baruchet, M.; et al. Src is activated by the nuclear receptor peroxisome proliferator-activated receptor β/δ in ultraviolet radiation-induced skin cancer. EMBO Mol. Med. 2014, 6, 80–98.
  51. Chong, H.C.; Tan, M.J.; Philippe, V.; Tan, S.H.; Tan, C.K.; Ku, C.W.; Goh, Y.Y.; Wahli, W.; Michalik, L.; Tan, N.S. Regulation of epithelial-mesenchymal IL-1 signaling by PPARbeta/delta is essential for skin homeostasis and wound healing. J. Cell Biol. 2009, 184, 817–831.
  52. Shureiqi, I.; Jiang, W.; Zuo, X.; Wu, Y.; Stimmel, J.B.; Leesnitzer, L.M.; Morris, J.S.; Fan, H.-Z.; Fischer, S.M.; Lippman, S.M. The 15-lipoxygenase-1 product 13-S-hydroxyoctadecadienoic acid down-regulates PPAR-delta to induce apoptosis in colorectal cancer cells. Proc. Natl. Acad. Sci. USA 2003, 100, 9968–9973.
  53. Yang, L.; Olsson, B.; Pfeifer, D.; Jönsson, J.-I.; Zhou, Z.-G.; Jiang, X.; Fredriksson, B.-A.; Zhang, H.; Sun, X.-F. Knockdown of peroxisome proliferator-activated receptor-beta induces less differentiation and enhances cell-fibronectin adhesion of colon cancer cells. Oncogene 2010, 29, 516–526.
  54. Marin, H.E.; Peraza, M.A.; Billin, A.N.; Willson, T.M.; Ward, J.M.; Kennett, M.J.; Gonzalez, F.J.; Peters, J.M. Ligand activation of peroxisome proliferator-activated receptor beta inhibits colon carcinogenesis. Cancer Res. 2006, 66, 4394–4401.
  55. Santos, G.C.; Zielenska, M.; Prasad, M.; Squire, J.A. Chromosome 6p amplification and cancer progression. J. Clin. Pathol. 2007, 60, 1–7.
  56. Uhlen, M.; Oksvold, P.; Fagerberg, L.; Lundberg, E.; Jonasson, K.; Forsberg, M.; Zwahlen, M.; Kampf, C.; Wester, K.; Hober, S.; et al. Towards a knowledge-based Human Protein Atlas. Nat. Biotechnol. 2010, 28, 1248–1250.
  57. Berglund, L.; Björling, E.; Oksvold, P.; Fagerberg, L.; Asplund, A.; Szigyarto, C.A.-K.; Persson, A.; Ottosson, J.; Wernérus, H.; Nilsson, P.; et al. A genecentric Human Protein Atlas for expression profiles based on antibodies. Mol. Cell. Proteomics 2008, 7, 2019–2027.
  58. Uhlén, M.; Björling, E.; Agaton, C.; Szigyarto, C.A.-K.; Amini, B.; Andersen, E.; Andersson, A.-C.; Angelidou, P.; Asplund, A.; Asplund, C.; et al. A human protein atlas for normal and cancer tissues based on antibody proteomics. Mol. Cell. Proteomics 2005, 4, 1920–1932.
  59. Foreman, J.E.; Sharma, A.K.; Amin, S.; Gonzalez, F.J.; Peters, J.M. Ligand activation of peroxisome proliferator-activated receptor-beta/delta (PPARbeta/delta) inhibits cell growth in a mouse mammary gland cancer cell line. Cancer Lett. 2010, 288, 219–225.
  60. Kittler, R.; Zhou, J.; Hua, S.; Ma, L.; Liu, Y.; Pendleton, E.; Cheng, C.; Gerstein, M.; White, K.P. A comprehensive nuclear receptor network for breast cancer cells. Cell Rep. 2013, 3, 538–551.
  61. Stephen, R.L.; Gustafsson, M.C.U.; Jarvis, M.; Tatoud, R.; Marshall, B.R.; Knight, D.; Ehrenborg, E.; Harris, A.L.; Wolf, C.R.; Palmer, C.N.A. Activation of peroxisome proliferator-activated receptor delta stimulates the proliferation of human breast and prostate cancer cell lines. Cancer Res. 2004, 64, 3162–3170.
  62. Girroir, E.E.; Hollingshead, H.E.; Billin, A.N.; Willson, T.M.; Robertson, G.P.; Sharma, A.K.; Amin, S.; Gonzalez, F.J.; Peters, J.M. Peroxisome proliferator-activated receptor-beta/delta (PPARbeta/delta) ligands inhibit growth of UACC903 and MCF7 human cancer cell lines. Toxicology 2008, 243, 236–243.
  63. Yao, P.-L.; Morales, J.L.; Zhu, B.; Kang, B.-H.; Gonzalez, F.J.; Peters, J.M. Activation of peroxisome proliferator-activated receptor-β/δ (PPAR-β/δ) inhibits human breast cancer cell line tumorigenicity. Mol. Cancer Ther. 2014, 13, 1008–1017.
  64. Palkar, P.S.; Borland, M.G.; Naruhn, S.; Ferry, C.H.; Lee, C.; Sk, U.H.; Sharma, A.K.; Amin, S.; Murray, I.A.; Anderson, C.R.; et al. Cellular and pharmacological selectivity of the peroxisome proliferator-activated receptor-beta/delta antagonist GSK3787. Mol. Pharmacol. 2010, 78, 419–430.
  65. Schug, T.T.; Berry, D.C.; Toshkov, I.A.; Cheng, L.; Nikitin, A.Y.; Noy, N. Overcoming retinoic acid-resistance of mammary carcinomas by diverting retinoic acid from PPARbeta/delta to RAR. Proc. Natl. Acad. Sci. USA 2008, 105, 7546–7551.
  66. Kannan-Thulasiraman, P.; Seachrist, D.D.; Mahabeleshwar, G.H.; Jain, M.K.; Noy, N. Fatty acid-binding protein 5 and PPARbeta/delta are critical mediators of epidermal growth factor receptor-induced carcinoma cell growth. J. Biol. Chem. 2010, 285, 19106–19115.
  67. Borland, M.G.; Khozoie, C.; Albrecht, P.P.; Zhu, B.; Lee, C.; Lahoti, T.S.; Gonzalez, F.J.; Peters, J.M. Stable over-expression of PPARβ/δ and PPARγ to examine receptor signaling in human HaCaT keratinocytes. Cell Signal. 2011, 23, 2039–2050.
  68. Borland, M.G.; Foreman, J.E.; Girroir, E.E.; Zolfaghari, R.; Sharma, A.K.; Amin, S.; Gonzalez, F.J.; Ross, A.C.; Peters, J.M. Ligand activation of peroxisome proliferator-activated receptor-beta/delta inhibits cell proliferation in human HaCaT keratinocytes. Mol. Pharmacol. 2008, 74, 1429–1442.
  69. Wang, X.; Wang, G.; Shi, Y.; Sun, L.; Gorczynski, R.; Li, Y.-J.; Xu, Z.; Spaner, D.E. PPAR-delta promotes survival of breast cancer cells in harsh metabolic conditions. Oncogenesis 2016, 5, e232.
  70. Burdick, A.D.; Bility, M.T.; Girroir, E.E.; Billin, A.N.; Willson, T.M.; Gonzalez, F.J.; Peters, J.M. Ligand activation of peroxisome proliferator-activated receptor-beta/delta(PPARbeta/delta) inhibits cell growth of human N/TERT-1 keratinocytes. Cell Signal. 2007, 19, 1163–1171.
  71. Adhikary, T.; Brandt, D.T.; Kaddatz, K.; Stockert, J.; Naruhn, S.; Meissner, W.; Finkernagel, F.; Obert, J.; Lieber, S.; Scharfe, M.; et al. Inverse PPARβ/δ agonists suppress oncogenic signaling to the ANGPTL4 gene and inhibit cancer cell invasion. Oncogene 2013, 32, 5241–5252.
  72. Yin, Y.; Russell, R.G.; Dettin, L.E.; Bai, R.; Wei, Z.-L.; Kozikowski, A.P.; Kopelovich, L.; Glazer, R.I. Peroxisome proliferator-activated receptor delta and gamma agonists differentially alter tumor differentiation and progression during mammary carcinogenesis. Cancer Res. 2005, 65, 3950–3957.
  73. Pollock, C.B.; Yin, Y.; Yuan, H.; Zeng, X.; King, S.; Li, X.; Kopelovich, L.; Albanese, C.; Glazer, R.I. PPARδ activation acts cooperatively with 3-phosphoinositide-dependent protein kinase-1 to enhance mammary tumorigenesis. PLoS ONE 2011, 6, e16215.
  74. Yuan, H.; Lu, J.; Xiao, J.; Upadhyay, G.; Umans, R.; Kallakury, B.; Yin, Y.; Fant, M.E.; Kopelovich, L.; Glazer, R.I. PPARδ induces estrogen receptor-positive mammary neoplasia through an inflammatory and metabolic phenotype linked to mTOR activation. Cancer Res. 2013, 73, 4349–4361.
  75. Ghosh, M.; Ai, Y.; Narko, K.; Wang, Z.; Peters, J.M.; Hla, T. PPARdelta is pro-tumorigenic in a mouse model of COX-2-induced mammary cancer. Prostaglandins Other Lipid Mediat. 2009, 88, 97–100.
  76. Gimble, J.M.; Pighetti, G.M.; Lerner, M.R.; Wu, X.; Lightfoot, S.A.; Brackett, D.J.; Darcy, K.; Hollingsworth, A.B. Expression of peroxisome proliferator activated receptor mRNA in normal and tumorigenic rodent mammary glands. Biochem. Biophys. Res. Commun. 1998, 253, 813–817.
  77. Fajas, L.; Auboeuf, D.; Raspé, E.; Schoonjans, K.; Lefebvre, A.M.; Saladin, R.; Najib, J.; Laville, M.; Fruchart, J.C.; Deeb, S.; et al. The organization, promoter analysis, and expression of the human PPARgamma gene. J. Biol. Chem. 1997, 272, 18779–18789.
  78. Fang, S.; Livergood, M.C.; Nakagawa, P.; Wu, J.; Sigmund, C.D. Role of the Peroxisome Proliferator Activated Receptors in Hypertension. Circ. Res. 2021, 128, 1021–1039.
  79. Francque, S.; Szabo, G.; Abdelmalek, M.F.; Byrne, C.D.; Cusi, K.; Dufour, J.-F.; Roden, M.; Sacks, F.; Tacke, F. Nonalcoholic steatohepatitis: The role of peroxisome proliferator-activated receptors. Nat. Rev. Gastroenterol. Hepatol. 2021, 18, 24–39.
  80. Azhar, S. Peroxisome proliferator-activated receptors, metabolic syndrome and cardiovascular disease. Future Cardiol. 2010, 6, 657–691.
  81. Takeyama, K.; Kodera, Y.; Suzawa, M.; Kato, S. . Nihon Rinsho. 2000, 58, 357–363.
  82. Janani, C.; Ranjitha Kumari, B.D. PPAR gamma gene--a review. Diabetes Metab. Syndr. 2015, 9, 46–50.
  83. Hamblin, M.; Chang, L.; Fan, Y.; Zhang, J.; Chen, Y.E. PPARs and the cardiovascular system. Antioxid. Redox Signal. 2009, 11, 1415–1452.
  84. Li, Y.; Qi, Y.; Huang, T.H.W.; Yamahara, J.; Roufogalis, B.D. Pomegranate flower: A unique traditional antidiabetic medicine with dual PPAR-alpha/-gamma activator properties. Diabetes. Obes. Metab. 2008, 10, 10–17.
  85. Blaschke, F.; Caglayan, E.; Hsueh, W.A. Peroxisome proliferator-activated receptor gamma agonists: Their role as vasoprotective agents in diabetes. Endocrinol. Metab. Clin. N. Am. 2006, 35, 561–574.
  86. Rizos, C.V.; Kei, A.; Elisaf, M.S. The current role of thiazolidinediones in diabetes management. Arch. Toxicol. 2016, 90, 1861–1881.
  87. Kahn, C.R.; Chen, L.; Cohen, S.E. Unraveling the mechanism of action of thiazolidinediones. J. Clin. Investig. 2000, 106, 1305–1307.
  88. Hauner, H. The mode of action of thiazolidinediones. Diabetes. Metab. Res. Rev. 2002, 18 (Suppl. S2), S10–S15.
  89. Ghoochani, A.; Shabani, K.; Peymani, M.; Ghaedi, K.; Karamali, F.; Karbalaei, K.; Tanhaie, S.; Salamian, A.; Esmaeili, A.; Valian-Borujeni, S.; et al. The influence of peroxisome proliferator-activated receptor γ(1) during differentiation of mouse embryonic stem cells to neural cells. Differentiation 2012, 83, 60–67.
  90. Farshbaf, M.J.; Ghaedi, K.; Shirani, M.; Nasr-Esfahani, M.H. Peroxisome proliferator activated receptor gamma (PPARγ) as a therapeutic target for improvement of cognitive performance in Fragile-X. Med. Hypotheses 2014, 82, 291–294.
  91. Yang, Y.; Zhao, L.-H.; Huang, B.; Wang, R.-Y.; Yuan, S.-X.; Tao, Q.-F.; Xu, Y.; Sun, H.-Y.; Lin, C.; Zhou, W.-P. Pioglitazone, a PPARγ agonist, inhibits growth and invasion of human hepatocellular carcinoma via blockade of the rage signaling. Mol. Carcinog. 2015, 54, 1584–1595.
  92. Wang, Y.; Tan, H.; Xu, D.; Ma, A.; Zhang, L.; Sun, J.; Yang, Z.; Liu, Y.; Shi, G. The combinatory effects of PPAR-γ agonist and survivin inhibition on the cancer stem-like phenotype and cell proliferation in bladder cancer cells. Int. J. Mol. Med. 2014, 34, 262–268.
  93. Velmurugan, B.K.; Yang, H.-H.; Sung, P.-J.; Weng, C.-F. Excavatolide B inhibits nonsmall cell lung cancer proliferation by altering peroxisome proliferator activated receptor gamma expression and PTEN/AKT/NF-Kβ expression. Environ. Toxicol. 2017, 32, 290–301.
  94. Srivastava, N.; Kollipara, R.K.; Singh, D.K.; Sudderth, J.; Hu, Z.; Nguyen, H.; Wang, S.; Humphries, C.G.; Carstens, R.; Huffman, K.E.; et al. Inhibition of cancer cell proliferation by PPARγ is mediated by a metabolic switch that increases reactive oxygen species levels. Cell Metab. 2014, 20, 650–661.
  95. Pestereva, E.; Kanakasabai, S.; Bright, J.J. PPARγ agonists regulate the expression of stemness and differentiation genes in brain tumour stem cells. Br. J. Cancer 2012, 106, 1702–1712.
  96. Trindade-da-Silva, C.A.; Reis, C.F.; Vecchi, L.; Napimoga, M.H.; Sperandio, M.; Matias Colombo, B.F.; Alves, P.T.; Ward, L.S.; Ueira-Vieira, C.; Goulart, L.R. 15-Deoxy-Δ(12,14)-prostaglandin J2 Induces Apoptosis and Upregulates SOCS3 in Human Thyroid Cancer Cells. PPAR Res. 2016, 2016, 4106297.
  97. Wu, K.; Yang, Y.; Liu, D.; Qi, Y.; Zhang, C.; Zhao, J.; Zhao, S. Activation of PPARγ suppresses proliferation and induces apoptosis of esophageal cancer cells by inhibiting TLR4-dependent MAPK pathway. Oncotarget 2016, 7, 44572–44582.
  98. Zurlo, D.; Ziccardi, P.; Votino, C.; Colangelo, T.; Cerchia, C.; Dal Piaz, F.; Dallavalle, S.; Moricca, S.; Novellino, E.; Lavecchia, A.; et al. The antiproliferative and proapoptotic effects of cladosporols A and B are related to their different binding mode as PPARγ ligands. Biochem. Pharmacol. 2016, 108, 22–35.
  99. Weidner, C.; Rousseau, M.; Micikas, R.J.; Fischer, C.; Plauth, A.; Wowro, S.J.; Siems, K.; Hetterling, G.; Kliem, M.; Schroeder, F.C.; et al. Amorfrutin C Induces Apoptosis and Inhibits Proliferation in Colon Cancer Cells through Targeting Mitochondria. J. Nat. Prod. 2016, 79, 2–12.
  100. Kohno, H.; Yasui, Y.; Suzuki, R.; Hosokawa, M.; Miyashita, K.; Tanaka, T. Dietary seed oil rich in conjugated linolenic acid from bitter melon inhibits azoxymethane-induced rat colon carcinogenesis through elevation of colonic PPARgamma expression and alteration of lipid composition. Int. J. Cancer 2004, 110, 896–901.
  101. Yasui, Y.; Hosokawa, M.; Sahara, T.; Suzuki, R.; Ohgiya, S.; Kohno, H.; Tanaka, T.; Miyashita, K. Bitter gourd seed fatty acid rich in 9c,11t,13t-conjugated linolenic acid induces apoptosis and up-regulates the GADD45, p53 and PPARgamma in human colon cancer Caco-2 cells. Prostaglandins. Leukot. Essent. Fatty Acids 2005, 73, 113–119.
  102. Mueller, E.; Sarraf, P.; Tontonoz, P.; Evans, R.M.; Martin, K.J.; Zhang, M.; Fletcher, C.; Singer, S.; Spiegelman, B.M. Terminal differentiation of human breast cancer through PPAR gamma. Mol. Cell 1998, 1, 465–470.
  103. Suh, N.; Wang, Y.; Williams, C.R.; Risingsong, R.; Gilmer, T.; Willson, T.M.; Sporn, M.B. A new ligand for the peroxisome proliferator-activated receptor-gamma (PPAR-gamma), GW7845, inhibits rat mammary carcinogenesis. Cancer Res. 1999, 59, 5671–5673.
  104. Moon, H.-S.; Guo, D.-D.; Lee, H.-G.; Choi, Y.-J.; Kang, J.-S.; Jo, K.; Eom, J.-M.; Yun, C.-H.; Cho, C.-S. Alpha-eleostearic acid suppresses proliferation of MCF-7 breast cancer cells via activation of PPARgamma and inhibition of ERK 1/2. Cancer Sci. 2010, 101, 396–402.
  105. Bonofiglio, D.; Gabriele, S.; Aquila, S.; Catalano, S.; Gentile, M.; Middea, E.; Giordano, F.; Andò, S. Estrogen receptor alpha binds to peroxisome proliferator-activated receptor response element and negatively interferes with peroxisome proliferator-activated receptor gamma signaling in breast cancer cells. Clin. Cancer Res. 2005, 11, 6139–6147.
  106. Bonofiglio, D.; Aquila, S.; Catalano, S.; Gabriele, S.; Belmonte, M.; Middea, E.; Qi, H.; Morelli, C.; Gentile, M.; Maggiolini, M.; et al. Peroxisome proliferator-activated receptor-gamma activates p53 gene promoter binding to the nuclear factor-kappaB sequence in human MCF7 breast cancer cells. Mol. Endocrinol. 2006, 20, 3083–3092.
  107. Bonofiglio, D.; Gabriele, S.; Aquila, S.; Qi, H.; Belmonte, M.; Catalano, S.; Andò, S. Peroxisome proliferator-activated receptor gamma activates fas ligand gene promoter inducing apoptosis in human breast cancer cells. Breast Cancer Res. Treat. 2009, 113, 423–434.
  108. Chinnaiyan, A.M.; Dixit, V.M. Portrait of an executioner: The molecular mechanism of FAS/APO-1-induced apoptosis. Semin. Immunol. 1997, 9, 69–76.
  109. Debatin, K.-M. Apoptosis pathways in cancer and cancer therapy. Cancer Immunol. Immunother. 2004, 53, 153–159.
  110. Pinkoski, M.J.; Green, D.R. Fas ligand, death gene. Cell Death Differ. 1999, 6, 1174–1181.
  111. Catalano, S.; Mauro, L.; Bonofiglio, D.; Pellegrino, M.; Qi, H.; Rizza, P.; Vizza, D.; Bossi, G.; Andò, S. In vivo and in vitro evidence that PPARγ ligands are antagonists of leptin signaling in breast cancer. Am. J. Pathol. 2011, 179, 1030–1040.
  112. Aronica, S.M.; Kraus, W.L.; Katzenellenbogen, B.S. Estrogen action via the cAMP signaling pathway: Stimulation of adenylate cyclase and cAMP-regulated gene transcription. Proc. Natl. Acad. Sci. USA 1994, 91, 8517–8521.
  113. Migliaccio, A.; Di Domenico, M.; Castoria, G.; de Falco, A.; Bontempo, P.; Nola, E.; Auricchio, F. Tyrosine kinase/p21ras/MAP-kinase pathway activation by estradiol-receptor complex in MCF-7 cells. EMBO J. 1996, 15, 1292–1300.
  114. Keller, H.; Givel, F.; Perroud, M.; Wahli, W. Signaling cross-talk between peroxisome proliferator-activated receptor/retinoid X receptor and estrogen receptor through estrogen response elements. Mol. Endocrinol. 1995, 9, 794–804.
  115. Cui, Y.; Miyoshi, K.; Claudio, E.; Siebenlist, U.K.; Gonzalez, F.J.; Flaws, J.; Wagner, K.-U.; Hennighausen, L. Loss of the peroxisome proliferation-activated receptor gamma (PPARgamma ) does not affect mammary development and propensity for tumor formation but leads to reduced fertility. J. Biol. Chem. 2002, 277, 17830–17835.
  116. Yin, Y.; Yuan, H.; Zeng, X.; Kopelovich, L.; Glazer, R.I. Inhibition of peroxisome proliferator-activated receptor gamma increases estrogen receptor-dependent tumor specification. Cancer Res. 2009, 69, 687–694.
  117. Saez, E.; Rosenfeld, J.; Livolsi, A.; Olson, P.; Lombardo, E.; Nelson, M.; Banayo, E.; Cardiff, R.D.; Izpisua-Belmonte, J.C.; Evans, R.M. PPAR gamma signaling exacerbates mammary gland tumor development. Genes Dev. 2004, 18, 528–540.
  118. Tian, L.; Zhou, J.; Casimiro, M.C.; Liang, B.; Ojeifo, J.O.; Wang, M.; Hyslop, T.; Wang, C.; Pestell, R.G. Activating peroxisome proliferator-activated receptor gamma mutant promotes tumor growth in vivo by enhancing angiogenesis. Cancer Res. 2009, 69, 9236–9244.
  119. Xu, Y.-Y.; Liu, H.; Su, L.; Xu, N.; Xu, D.-H.; Liu, H.-Y.; Spaner, D.; Bed-David, Y.; Li, Y.-J. PPARγ inhibits breast cancer progression by upregulating PTPRF expression. Eur. Rev. Med. Pharmacol. Sci. 2019, 23, 9965–9977.
  120. Kotta-Loizou, I.; Giaginis, C.; Theocharis, S. The role of peroxisome proliferator-activated receptor-γ in breast cancer. Anticancer. Agents Med. Chem. 2012, 12, 1025–1044.
  121. Yang, P.-B.; Hou, P.-P.; Liu, F.-Y.; Hong, W.-B.; Chen, H.-Z.; Sun, X.-Y.; Li, P.; Zhang, Y.; Ju, C.-Y.; Luo, L.-J.; et al. Blocking PPARγ interaction facilitates Nur77 interdiction of fatty acid uptake and suppresses breast cancer progression. Proc. Natl. Acad. Sci. USA 2020, 117, 27412–27422.
  122. Papadaki, I.; Mylona, E.; Giannopoulou, I.; Markaki, S.; Keramopoulos, A.; Nakopoulou, L. PPARgamma expression in breast cancer: Clinical value and correlation with ERbeta. Histopathology 2005, 46, 37–42.
  123. Kulkarni, S.; Patil, D.B.; Diaz, L.K.; Wiley, E.L.; Morrow, M.; Khan, S.A. COX-2 and PPARgamma expression are potential markers of recurrence risk in mammary duct carcinoma in-situ. BMC Cancer 2008, 8, 36.
  124. Liedtke, C.; Mazouni, C.; Hess, K.R.; André, F.; Tordai, A.; Mejia, J.A.; Symmans, W.F.; Gonzalez-Angulo, A.M.; Hennessy, B.; Green, M.; et al. Response to neoadjuvant therapy and long-term survival in patients with triple-negative breast cancer. J. Clin. Oncol. 2008, 26, 1275–1281.
  125. Li, T.; Zhang, Q.; Zhang, J.; Yang, G.; Shao, Z.; Luo, J.; Fan, M.; Ni, C.; Wu, Z.; Hu, X. Fenofibrate induces apoptosis of triple-negative breast cancer cells via activation of NF-κB pathway. BMC Cancer. 2014, 14, 96.
  126. Kwong, S.C.; Jamil, A.H.A.; Rhodes, A.; Taib, N.A.; Chung, I. Metabolic role of fatty acid binding protein 7 in mediating triple-negative breast cancer cell death via PPAR-α signaling. J. Lipid Res. 2019, 60, 1807–1817.
  127. Bonofiglio, D.; Cione, E.; Qi, H.; Pingitore, A.; Perri, M.; Catalano, S.; Vizza, D.; Panno, M.L.; Genchi, G.; Fuqua, S.A.W.; et al. Combined low doses of PPARgamma and RXR ligands trigger an intrinsic apoptotic pathway in human breast cancer cells. Am. J. Pathol. 2009, 175, 1270–1280.
  128. Apaya, M.K.; Hsiao, P.W.; Yang, Y.C.; Shyur, L.F. Deregulating the CYP2C19/Epoxy-Eicosatrienoic Acid-Associated FABP4/FABP5 Signaling Network as a Therapeutic Approach for Metastatic Triple-Negative Breast Cancer. Cancers 2020, 12, 199.
More
Information
Subjects: Cell Biology
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , , ,
View Times: 323
Revisions: 2 times (View History)
Update Date: 24 Feb 2023
1000/1000
Video Production Service