Skip to main content

MINI REVIEW article

Front. Pharmacol., 21 March 2023
Sec. Pharmacology of Anti-Cancer Drugs

Peroxisome proliferator-activated receptors regulate the progression and treatment of gastrointestinal cancers

  • 1Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, China
  • 2Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
  • 3Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
  • 4Hubei Engineering Research Center for Immunological Diagnosis and Therapy of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China

Peroxisome proliferator-activated receptors (PPARs) are essential nuclear hormone receptors regulating metabolic processes, and they participate in the initiation and progression processes of tumors. Gastrointestinal (GI) cancer is a prevalent malignancy worldwide that originates from the tissues of the gastrointestinal tract and is characterized by severe symptoms and poor prognosis. Numerous published studies have investigated the critical role of PPARs in esophageal, gastric, and colorectal cancers. Here, we summarize and review the current literature to understand the role of PPARs in the pathogenesis of GI cancers and to provide a systematic reference for the subsequent investigation and development of efficient therapies targeting PPARs and their pathways.

Introduction

Gastrointestinal (GI) cancer is a common malignancy worldwide that originates from the gastrointestinal tract and has the characteristics of severe symptoms and poor prognosis (Cui et al., 2021). The GI tract is the most important component of the digestive system, with the functions of food digestion and nutrition absorption, as well as an innate immune system for health monitoring (Fenneman et al., 2023). Malignant transition of the normal of GI tract not only affects the efficiency of nutrition absorption but also causes secondary dysfunction in other organs once metastasis occurs. Specifically, GI cancers include cancers originating from the esophagus, stomach, and colorectum, which together account for about 15 percent of all kinds of cancer (Sung et al., 2021). Thus, it is important to clarify the pathogenesis mechanisms in the progression of GI cancers, which could help provide precise targets for developing efficient therapeutics.

Peroxisome proliferator-activated receptors (PPARs) are nuclear hormone receptors activated by fatty acids to regulate the metabolic processes, with three separate isotypes known as PPARα, PPARγ, and PPARβ/δ (Christofides et al., 2021). Although the three subtypes of PPARs can sense nutritional status and regulate the energy metabolism process, their mechanisms vary significantly. PPARα can regulate the expression of essential enzymes and proteins by regulating the transcriptional process of targeted genes, which further participate in the lipid transportation and β-oxidation of fatty acids, resulting in fat degradation (Rigano et al., 2017). PPARγ is widely expressed in different tissues and organs, governing the expression of essential genes using two isoforms of PPARγ, with different lengths, to be activated by agonists (Neschen et al., 2007; Bernardo et al., 2009; Chen et al., 2012; Janani and Ranjitha Kumari, 2015). PPARδ, another isotype, is highly expressed in tissues such as the lung, kidney, and heart, and can regulate the expression of target genes involved in cardiovascular diseases, cancers, and gastrointestinal diseases in combination with progression and treatment at the DNA-binding domain (Ratziu et al., 2016; Kadayat et al., 2020; Xiao and Wang, 2022).

The metabolic processes regulated by PPARs are very important to the function of various organs, and the disturbance of PPARs and related pathways could lead to tumorigenesis, especially in the parts of the gastrointestinal system where nutrition is absorbed. In recent years, many researchers have focused on investigating the role of PPARs in GI cancers and on developing efficient targeted therapeutics (Wu et al., 2016; Xiao and Wang, 2022; Fenneman et al., 2023). In 2012, a team from Italy led by Vittorio Colantuoni systematically summarized and reviewed the roles of PPARs in GI cancers (Fucci et al., 2012). Thus, it is necessary to conclude and summarize the recent published works in the field of PPARs and GI cancers to systematically reveal the implications of PPARs for cancer diagnostics and treatment.

Regulatory role of PPARs in esophageal cancers

Esophageal cancer (EC) is a prevalent malignant tumor occurring in the mucosal epithelium of the esophagus and includes two main kinds: squamous cell carcinoma (ESCC) and adenocarcinoma (Short et al., 2017). The cancer’s recently published statistical data show that its survival rate is still very low, about 20%, although the overall survival rate for all cancers has increased (Siegel et al., 2022). Unhealthy dietary habits including hot up taking, nutrition deficiency, and alcohol consumption are regarded as essential risk factors for ESCC (van den Brandt, 2022). Risk factors for EC mainly include Barrett’s esophagus, obesity, smoking, and gastroesophageal reflux disease (Lordick et al., 2016; Lagergren et al., 2017). Surgery, chemotherapy, and chemoradiotherapy are the three traditional EC therapies. Neoadjuvant chemoradiotherapy or chemotherapy before surgery is recommended for patients with metastases or at more advanced stages, or who are not suitable for surgery. However, most patients suffer from relapse and poor prognosis (Shah et al., 2020). Therefore, it is urgent to develop more novel and efficient therapeutic tactics for esophageal cancer treatment. With the proposed conception of precise medicine, more and more investigations are exploring the regulatory mechanisms of EC progression, especially the regulatory functions and roles of PPARs.

The roles of PPARs in regulating esophageal cancers

Yue et al. analyzed esophageal cancer microarray data to profile microRNA expression and construct networks of differentially expressed genes and microRNAs using a bioinformatics approach. The subsequent gene enrichment analysis identified ADCYAP1R1, in the PPAR signaling pathway, as potentially involved in the prognosis of esophageal cancer (Chen et al., 2022a). A study conducted by Yukio Terashita et al. examined the correlation between the mRNA expression of PPARγ and esophageal cancer prognosis in 55 patients with primary ESCCs and found that PPARγ expression decreased significantly compared with the normal esophageal epithelium; thus, they concluded that the mRNA expression level of PPARγ could be regarded as a prognosis marker for esophageal cancer patients (Terashita et al., 2002).

Strategies targeting PPARs to treat esophageal cancers

Targeting the complex regulatory mechanisms of PPARs could provide more opportunities for treatment of ECs. A study investigated the expression profiles of PPARγ in ESCCs and used a novel PPARγ agonist, efatutazone, to treat ESCC cells. The investigators found that the expression of PPARγ was inversely related with Ki-67, and its expression was associated independently with good prognosis in ESCC. In addition, application of efatutazone could inhibit the proliferation of ESCC cells in vitro and in vivo (Sawayama et al., 2014). Furthermore, a team used the PPARγ agonist rosiglitazone (RGZ) to treat esophageal cancer cells and discovered that application of RGZ could suppress cellular proliferation and induce apoptosis, while PPARγ inhibition could partially restore the RGZ-treated effect, suggesting that activation of PPARγ with specific agonists could inhibit cancer cell growth and cancer progression (Wu et al., 2016). In addition to PPARγ, PPARδ could also act as an essential mediator for 83b1 to exert anti-cancer effects on human esophageal cancers by down-regulating cancer-related genes and molecules (Pun et al., 2017).

Regulatory role of PPARs in gastric cancer progression and treatment

Gastric cancer (GC) is a severe malignancy and the fifth most common cancer, with about a million new patients suffering this disease every year. It is commonly diagnosed in the late stage, with rapid progression; therefore, the mortality rate remains high despite huge progress in diagnostic equipment and therapies, and it remains the third deadliest cancer in the world (Ferlay et al., 2019). During the proliferation of cancer cells and the progression and metastasis of tumors, large amounts of nutrition and energy are required to support tumorigenesis. PPARs serve as metabolic regulatory mediators, generating energy in response to nutrition stimulation, and play critical roles in the progression of GC. Recently, more and more investigations have focused on the regulatory mechanisms of PPARs, broadening the mechanism networks of GC development (Chen et al., 2022b).

The roles of PPARs in regulating gastric cancers

A study performed by Yu-Ching et al. investigated the role of visceral-to-subcutaneous fat ratio in GC and found expression of the three isotypes of PPARs to be decreased in patients with higher ratios of visceral adipose tissue versus subcutaneous adipose tissue, compared to ones with lower ratios. Furthermore, adipocytes co-cultured with cancer cells could accelerate their proliferation by downregulating the expression of PPARs (Lin et al., 2021). Additionally, recent research has revealed PPAR pathways to be inhibited in patients with gastric carcinoma of the T300A/T300A genotype, accompanied by an increased ratio of tumor cell apoptosis and overall patient survival improvement (Ma et al., 2021). One study investigating the role of N-myc downstream regulated gene 1 (NDRG1) expression in GCs revealed that the downregulation of NDRG1 indicated malignant behaviors and a poor prognosis, and clarified their relationship with the PPAR signaling pathways (Xiao and Zheng, 2020). Moreover, lncRNA HCP5 derived from MSC could promote the oxidation of fatty acids in gastric cancer cells, thus increasing their stemness and chemo-resistance via regulating the signaling axis of miR-3619-5p/AMPK/CEBPB mediated by PPARγ coactivator 1A, demonstrating a novel target of HCP5 as an efficient therapy for GCs (Wu et al., 2020). A team investigating adenomas in early gastric cancer of low-grade dysplasia and high-grade dysplasia analyzed their genomic and transcriptomic landscape and revealed the signaling pathway of PPARs to be decreased significantly in early gastric cancer, while the pathways associated with extracellular matrix and focal adhesion were upregulated (Min et al., 2016). Christie Jeon et al. performed an analysis of the SNPs in PPARs with gastric cancer to illuminate the association between each genotype and gastric cancer and found that PPARδ variants were significantly related to gastric cancer, implicating a role for drugs targeting PPARδ in preventing the progression of gastric cancer (Jeon et al., 2013). In gastric mucosal cells, walnut polyphenol extracts (WPEs) induced nuclear translocation of PPARγ and significantly attenuated the gastric pathologies induced by HP (Park et al., 2020). Similarly, in gastric epithelial cells infected with Helicobacter pylori (HP), β-carotene could prevent cancer cell invasion by inhibiting the expression of MMP-10 mediated by mitogen-activated protein kinase (MAPK), which is due to increased expression of PPARγ-mediated catalase and reduced levels of ROS (Bae et al., 2021).

Strategies targeting PPARs to treat gastric cancers

With deep research on the regulatory role of PPARs in gastric cancer, more and more investigations are identifying precise therapies for gastric cancer treatment. Fang Guo et al. revealed that cellular proliferation and migration in GC could be inhibited by PPARγ mediating the suppression of TERT and ENAH, implying an important regulatory role for PPARγ and providing solid potential targets for GC therapy (Guo et al., 2016). Another study published recently confirmed that the expression of fatty acid-binding protein 4 (FABP4) in GC patients is positively related to poor outcomes. FABP4 could regulate the translocation of PPARγ to promote the subsequent transcription and expression of cell adhesion molecule 3 (CADM3), which could further inhibit GC metastasis. In addition, application of the PPARγ agonist rosiglitazone could directly activate the downstream effect of FABP4, regulating CADM3 to inhibit the metastasis of GC cells in vivo and in vitro (Chen et al., 2022b). Meanwhile, a research group reported that the upregulation of PPARα in gastric cancers is negatively correlated with prognosis and that treatment with fenofibrate could regulate PPARα pathways to reverse the dysfunction of mitochondria and cellular metabolism (Chen et al., 2020). In immunotherapy for gastric cancer, PPARγ could induce the expression of DOK1 in macrophages and empower the conversion of macrophages to the inflammatory phenotype, thus inhibiting the viability of gastric cancer cells (Li et al., 2019). A group of researchers tried to develop novel therapeutic drugs targeting PPARs. Using screening methods, Keisuke Yamamoto et al. designed a novel class of PPARγ ligand 1, which is more potent in gastric cancer cells, with a dibenzoazepine scaffold (Yamamoto et al., 2018). Masao Ohashi et al. developed hPPARγ-selective agonist 3 and improved its aqueous solubility to facilitate its application to gastric cancer (Ohashi et al., 2013). In conclusion, these published studies reveal the broad application of PPARs for gastric cancer treatment.

Regulatory roles of PPARs in colorectal cancer progression and treatment

Colorectal cancer (CRC) is a common malignant tumor of the digestive tract and accounts for the second highest mortality among cancers (Sung et al., 2021). Global health burden analysis of cancers has shown the incidence rate of colorectal cancer to be about 10.2% and the mortality rate to be about 9.2%, which make it a significant global burden. The pathogenesis of colorectal cancer is derived from the uncontrolled proliferation of epithelial cells, which could further destroy the mucosa and invade the deeper tissues, thus affecting the integrity of the colorectal structure (Terzić et al., 2010). Currently, surgery, chemotherapy, and radiotherapy are considered the main treatments for colon cancer, and the choice of treatment relies on the stage of cancer and the specific location. Generally, surgery is suitable for cancers at early stages, while chemotherapy and radiotherapy are applicable for later-stage cancers (Peters, 2019). Furthermore, due to the latent symptoms of CRC, it is common for patients to be diagnosed with CRC at a late stage, which is the leading cause of the high mortality of this disease (Shi et al., 2021). Therefore, it is necessary to pave new avenues for the treatment of CRC by investigating its pathogenesis mechanisms.

Roles of PPARs in regulating colorectal cancers

Systematic analysis of PPARs by bioinformatics has identified the critical roles of PPARs in the progression and prognosis of CRC. Md Misbah et al. investigated the mechanisms of drug resistance and identified biomarkers of CRC in colon cancer cell lines, revealing PPAR signaling to be one of the pathways enriched by different expression genes (Misbah et al., 2022). In addition, Ozlem Kurnaz et al. confirmed that there is increased risk for patients with the PPARγ T161 allele in CRC development (Kurnaz-Gomleksiz et al., 2022). Additionally, bioinformatics methods were employed to identify genes with different expressions in CRC, and PPAR signaling pathways were demonstrated to play an essential role in the occurrence of colon cancer (Zhang et al., 2022). Regarding the metastatic property of CRC, PPAR signaling pathways were shown to be significantly enriched in metastasis mechanisms by integrating analysis (Liu et al., 2021). Recently, single-cell analysis of primary and metastatic colorectal cancer tumors showed the PPAR signaling pathway to be enriched in the regulatory mechanisms of CRC tumorigenesis and metastasis (Wang et al., 2022).

PPAR signaling pathways play critical roles in colorectal cancer progression. The FABP4 and PPARγ axis has been demonstrated to be a novel target for CRC, while PPARδ has been identified as a tumor suppressor in CRC cells (Zhou et al., 2014; Zhao et al., 2019). More studies have revealed that PPARγ can function as a tumor suppressor in CRC cells via regulating miR-145 (Panza et al., 2014) and that PPARγ could induce the expression of PD-L1 in MSS+ CRC cells, for which immunogenicity can be enhanced by the pharmacological application of PPARγ agonists (Gutting et al., 2021). Teresa Friedrich et al. studied the role of docking protein 1 (DOK1) in CRC and found PPARγ to be one of the subsequent effectors to mediate the inhibitory effect of proliferation (Friedrich et al., 2016).

Strategies targeting PPARs to treat colorectal cancers

Apart from the pathogenic role of PPARs, numerous therapies aiming at PPAR signaling pathways have emerged recently. A team systematically analyzed different genes from different colorectal cancer cell lines and identified PPAR as an important indicator affecting radiotherapy sensitivity (Chen et al., 2017). Alessandra Ammazzalorso et al. synthesized novel benzothiazole amides 2b to stimulate the activity of PPARα in order to inhibit the proliferation of CRC cells (2019). N-Acylethanolamine acid amidase (NAAA) was upregulated in CRC patients, and inhibition of NAAA could reduce the proliferation of CRC cells mediated by PPARα (Romano et al., 2022). Moreover, several traditional Chinese drugs were demonstrated to be efficient in the treatment of colorectal cancer. Luteolin could be used as an agonist to activate PPARγ, which can upregulate organic cation/carnitine transporter 2 (OCTN2) to enhance the sensitivity of radiology (Qu et al., 2014); buffalo milk whey was also revealed to have an anti-tumor role through activating the necroptotic pathway and apoptotic pathways, accompanied by the upregulation of PPARγ and the downregulation of PPARα (Cacciola et al., 2022). In conclusion, these published studies systematically revealed the regulatory roles of PPARs in colorectal cancer progression and treatment.

Conclusion

Genetic regulation, environmental influence, and dietary habits comprehensively contribute to the pathogenesis of cancers. In gastrointestinal cancers, the key role of dietary habits is becoming increasingly important, not only by affecting the local environment of the gastrointestinal tract but also by influencing the entire nutrition metabolism of the body. PPARs are regarded as a kind of central regulatory factor in metabolism and energy conversion, and they regulate diverse aspects of cancer initiation, progression, and prognosis. Overall, PPARs could play protective roles in the homeostasis and function of the gastrointestinal tract, and agonists used to activate PPARs could theoretically inhibit the progression of GI cancers. Some studies have discovered that PPARs could function as promoters of cancers, possibly due to the difference between cancer cells and the tumor stages. Although numerous studies have focused on the roles and mechanisms of PPARs, more studies are still needed to broaden the signaling network and provide more targets for the precise therapy.

Author contributions

MZ: conceptualization and original draft writing. SH: conceptualization, writing review, and editing.

Funding

This work is supported by the First Affiliated Hospital of Nanchang University Young Talents Research and Cultivation Project, Grant Number YFYPY202122.

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors, and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

Ammazzalorso, A., De Lellis, L., Florio, R., Laghezza, A., De Filippis, B., Fantacuzzi, M., et al. (2019). Synthesis of novel benzothiazole amides: Evaluation of PPAR activity and anti-proliferative effects in paraganglioma, pancreatic and colorectal cancer cell lines. Bioorg. Med. Chem. Lett. 29 (16), 2302–2306. doi:10.1016/j.bmcl.2019.06.020

PubMed Abstract | CrossRef Full Text | Google Scholar

Bae, S., Lim, J. W., and Kim, H. (2021). β-Carotene inhibits expression of matrix metalloproteinase-10 and invasion in Helicobacter pylori-infected gastric epithelial cells. Mol. (Basel, Switz. 26 (6), 1567. doi:10.3390/molecules26061567

CrossRef Full Text | Google Scholar

Bernardo, A., Bianchi, D., Magnaghi, V., and Minghetti, L. (2009). Peroxisome proliferator-activated receptor-gamma agonists promote differentiation and antioxidant defenses of oligodendrocyte progenitor cells. J. Neuropathology Exp. Neurology 68 (7), 797–808. doi:10.1097/NEN.0b013e3181aba2c1

CrossRef Full Text | Google Scholar

Cacciola, N. A., Salzano, A., D'Onofrio, N., Venneri, T., Cicco, P. D., Vinale, F., et al. (2022). Buffalo milk whey activates necroptosis and apoptosis in a xenograft model of colorectal cancer. Int. J. Mol. Sci. 23 (15), 8464. doi:10.3390/ijms23158464

PubMed Abstract | CrossRef Full Text | Google Scholar

Chen, H., Shi, X., Ren, L., Zhuo, H., Zeng, L., Qin, Q., et al. (2022). Identification of the miRNA-mRNA regulatory network associated with radiosensitivity in esophageal cancer based on integrative analysis of the TCGA and GEO data. BMC Med. Genomics 15 (1), 249. doi:10.1186/s12920-022-01392-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Chen, L., Peng, J., Wang, Y., Jiang, H., Wang, W., Dai, J., et al. (2020). Fenofibrate-induced mitochondrial dysfunction and metabolic reprogramming reversal: The anti-tumor effects in gastric carcinoma cells mediated by the PPAR pathway. Am. J. Transl. Res. 12 (2), 428–446.

PubMed Abstract | Google Scholar

Chen, L., Zhu, Z., Gao, W., Jiang, Q., Yu, J., and Fu, C. (2017). Systemic analysis of different colorectal cancer cell lines and TCGA datasets identified IGF-1R/EGFR-PPAR-CASPASE axis as important indicator for radiotherapy sensitivity. Gene 627, 484–490. doi:10.1016/j.gene.2017.07.003

PubMed Abstract | CrossRef Full Text | Google Scholar

Chen, Q.-Y., Huang, X.-B., Zhao, Y.-J., Wang, H. G., Wang, J. B., Liu, L. C., et al. (2022). The peroxisome proliferator-activated receptor agonist rosiglitazone specifically represses tumour metastatic potential in chromatin inaccessibility-mediated FABP4-deficient gastric cancer. Theranostics 12 (4), 1904–1920. doi:10.7150/thno.66814

PubMed Abstract | CrossRef Full Text | Google Scholar

Chen, Y.-C., Wu, J.-S., Tsai, H.-D., Huang, C. Y., and Sun, G. Y., (2012). Peroxisome proliferator-activated receptor gamma (PPAR-γ) and neurodegenerative disorders. Mol. Neurobiol. 46 (1), 114–124. doi:10.1007/s12035-012-8259-8

PubMed Abstract | CrossRef Full Text | Google Scholar

Christofides, A., Konstantinidou, E., Jani, C., and Boussiotis, V. A. (2021). The role of peroxisome proliferator-activated receptors (PPAR) in immune responses. Metabolism: Clin. Exp. 114, 154338. doi:10.1016/j.metabol.2020.154338

CrossRef Full Text | Google Scholar

Cui, C., Lan, P., and Fu, L. (2021). The role of myeloid-derived suppressor cells in gastrointestinal cancer. Cancer Commun. Lond. Engl. 41 (6), 442–471. doi:10.1002/cac2.12156

CrossRef Full Text | Google Scholar

Fenneman, A. C., Weidner, M., Chen, L. A., Nieuwdorp, M., and Blaser, M. J. (2023). Antibiotics in the pathogenesis of diabetes and inflammatory diseases of the gastrointestinal tract. Nat. Rev. Gastroenterology Hepatology 20 (2), 81–100. doi:10.1038/s41575-022-00685-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Ferlay, J., Colombet, M., Soerjomataram, I., Mathers, C., Parkin, D. M., Pineros, M., et al. (2019). Estimating the global cancer incidence and mortality in 2018: GLOBOCAN sources and methods. Int. J. Cancer 144 (8), 1941–1953. doi:10.1002/ijc.31937

PubMed Abstract | CrossRef Full Text | Google Scholar

Friedrich, T., Söhn, M., Gutting, T., Janssen, K. P., Behrens, H. M., Rocken, C., et al. (2016). Subcellular compartmentalization of docking protein-1 contributes to progression in colorectal cancer. EBioMedicine 8, 159–172. doi:10.1016/j.ebiom.2016.05.003

PubMed Abstract | CrossRef Full Text | Google Scholar

Fucci, A., Colangelo, T., Votino, C., Pancione, M., Sabatino, L., and Colantuoni, V. (2012). The role of peroxisome proliferator-activated receptors in the esophageal, gastric, and colorectal cancer. PPAR Res. 2012, 242498. doi:10.1155/2012/242498

PubMed Abstract | CrossRef Full Text | Google Scholar

Guo, F., Ren, X., Dong, Y., Hu, X., Xu, D., Zhou, H., et al. (2016). Constitutive expression of PPARγ inhibits proliferation and migration of gastric cancer cells and down-regulates Wnt/β-Catenin signaling pathway downstream target genes TERT and ENAH. Gene 584 (1), 31–37. doi:10.1016/j.gene.2016.03.003

PubMed Abstract | CrossRef Full Text | Google Scholar

Gutting, T., Hauber, V., Pahl, J., Klapproth, K., Wu, W., Dobrota, I., et al. (2021). PPARγ induces PD-L1 expression in MSS+ colorectal cancer cells. Oncoimmunology 10 (1), 1906500. doi:10.1080/2162402X.2021.1906500

PubMed Abstract | CrossRef Full Text | Google Scholar

Janani, C., and Ranjitha Kumari, B. D. (2015). PPAR gamma gene--a review. Diabetes and Metabolic Syndrome 9 (1), 46–50. doi:10.1016/j.dsx.2014.09.015

PubMed Abstract | CrossRef Full Text | Google Scholar

Jeon, C., Chang, S.-C., Mu, L., Zhao, J., Rao, J. Y., Lu, Q. Y., et al. (2013). Genetic variants of peroxisome proliferator-activated receptor δ are associated with gastric cancer. Dig. Dis. Sci. 58 (10), 2881–2886. doi:10.1007/s10620-013-2770-2

PubMed Abstract | CrossRef Full Text | Google Scholar

Kadayat, T. M., Shrestha, A., Jeon, Y. H., An, H., Kim, J., Cho, S. J., et al. (2020). Targeting peroxisome proliferator-activated receptor delta (PPARδ): A medicinal chemistry perspective. J. Med. Chem. 63 (18), 10109–10134. doi:10.1021/acs.jmedchem.9b01882

PubMed Abstract | CrossRef Full Text | Google Scholar

Kurnaz-Gomleksiz, O., Torun, B. C., Isbir, T., Bulut, T., Sokucu, N., Yilmaz-Aydogan, H., et al. (2022). The role of PPAR-gamma C161T polymorphism in colorectal cancer susceptibility. Vivo (Athens, Greece) 36 (4), 1911–1915. doi:10.21873/invivo.12911

CrossRef Full Text | Google Scholar

Lagergren, J., Smyth, E., Cunningham, D., and Lagergren, P. (2017). Oesophageal cancer. Lancet (London, Engl. 390 (10110), 2383–2396. doi:10.1016/S0140-6736(17)31462-9

CrossRef Full Text | Google Scholar

Li, T., Li, B., Sara, A., Ay, C., Leung, W. Y., Zhang, Y., et al. (2019). Docking protein-1 promotes inflammatory macrophage signaling in gastric cancer. Oncoimmunology 8 (11), e1649961. doi:10.1080/2162402X.2019.1649961

PubMed Abstract | CrossRef Full Text | Google Scholar

Lin, Y.-C., Lin, G., and Yeh, T.-S. (2021). Visceral-to-subcutaneous fat ratio independently predicts the prognosis of locally advanced gastric cancer------ highlighting the role of adiponectin receptors and PPARα, β/δ, ɤ. Eur. J. Surg. Oncol. 47 (12), 3064–3073. doi:10.1016/j.ejso.2021.04.028

PubMed Abstract | CrossRef Full Text | Google Scholar

Liu, S., Zhang, Y., Zhang, S., Qiu, L., Zhang, B., and Han, J. (2021). Identification of hub genes related to liver metastasis of colorectal cancer by integrative analysis. Front. Oncol. 11, 714866. doi:10.3389/fonc.2021.714866

PubMed Abstract | CrossRef Full Text | Google Scholar

Lordick, F., Mariette, C., Haustermans, K., Obermannova, R., and Arnold, D. (2016). Oesophageal cancer: ESMO clinical practice guidelines for diagnosis, treatment and follow-up. Ann. Oncol. 27 (5), v50–v57. doi:10.1093/annonc/mdw329

PubMed Abstract | CrossRef Full Text | Google Scholar

Ma, C., Storer, C. E., Chandran, U., LaFramboise, W. A., Petrosko, P., Frank, M., et al. (2021). Crohn's disease-associated ATG16L1 T300A genotype is associated with improved survival in gastric cancer. EBioMedicine 67, 103347. doi:10.1016/j.ebiom.2021.103347

PubMed Abstract | CrossRef Full Text | Google Scholar

Min, B.-H., Hwang, J., Kim, N. K., Park, G., Kang, S. Y., Ahn, S., et al. (2016). Dysregulated Wnt signalling and recurrent mutations of the tumour suppressor RNF43 in early gastric carcinogenesis. J. Pathology 240 (3), 304–314. doi:10.1002/path.4777

CrossRef Full Text | Google Scholar

Misbah, M., Kumar, M., Lee, K.-H., and Shen, S. C. (2022). Identification of novel miRNAs, targeting genes, signaling pathway, and the small molecule for overcoming oxaliplatin resistance of metastatic colorectal cancer. BioMed Res. Int. 2022, 3825760. doi:10.1155/2022/3825760

PubMed Abstract | CrossRef Full Text | Google Scholar

Neschen, S., Morino, K., Dong, J., Wang-Fischer, Y., Cline, G. W., Romanelli, A. J., et al. (2007). n-3 Fatty acids preserve insulin sensitivity in vivo in a peroxisome proliferator-activated receptor-alpha-dependent manner. Diabetes 56 (4), 1034–1041. doi:10.2337/db06-1206

PubMed Abstract | CrossRef Full Text | Google Scholar

Ohashi, M., Oyama, T., Putranto, E. W., Waku, T., Nobusada, H., Kataoka, K., et al. (2013). Design and synthesis of a series of α-benzyl phenylpropanoic acid-type peroxisome proliferator-activated receptor (PPAR) gamma partial agonists with improved aqueous solubility. Bioorg. Med. Chem. 21 (8), 2319–2332. doi:10.1016/j.bmc.2013.02.003

PubMed Abstract | CrossRef Full Text | Google Scholar

Panza, A., Votino, C., Gentile, A., Valvano, M. R., Colangelo, T., Pancione, M., et al. (2014). Peroxisome proliferator-activated receptor γ-mediated induction of microRNA-145 opposes tumor phenotype in colorectal cancer. Biochimica Biophysica Acta 1843 (6), 1225–1236. doi:10.1016/j.bbamcr.2014.03.003

CrossRef Full Text | Google Scholar

Park, J. M., An, J. M., Han, Y. M., Surh, Y. J., Hwang, S. J., Kim, S. J., et al. (2020). Walnut polyphenol extracts inhibit Helicobacter pylori-induced STAT3Tyr705 phosphorylation through activation of PPAR-γ and SOCS1 induction. J. Clin. Biochem. Nutr. 67 (3), 248–256. doi:10.3164/jcbn.20-89

PubMed Abstract | CrossRef Full Text | Google Scholar

Peters, W. R. (2019). What every colorectal surgeon should know about the new American cancer society's colorectal cancer screening guidelines. Dis. Colon Rectum 62 (4), 397–398. doi:10.1097/DCR.0000000000001302

PubMed Abstract | CrossRef Full Text | Google Scholar

Pun, I. H. Y., Chan, D., Chan, S. H., Chung, P. Y., Zhou, Y. Y., Law, S., et al. (2017). Anti-cancer effects of a novel quinoline derivative 83b1 on human esophageal squamous cell carcinoma through down-regulation of COX-2 mRNA and PGE2. Cancer Res. Treat. 49 (1), 219–229. doi:10.4143/crt.2016.190

PubMed Abstract | CrossRef Full Text | Google Scholar

Qu, Q., Qu, J., Guo, Y., Zhou, B. T., and Zhou, H. H. (2014). Luteolin potentiates the sensitivity of colorectal cancer cell lines to oxaliplatin through the PPARγ/OCTN2 pathway. Anti-cancer Drugs 25 (9), 1016–1027. doi:10.1097/CAD.0000000000000125

PubMed Abstract | CrossRef Full Text | Google Scholar

Ratziu, V., Harrison, S. A., Francque, S., Bedossa, P., Lehert, P., Serfaty, L., et al. (2016). Elafibranor, an agonist of the peroxisome proliferator-activated receptor-α and -δ, induces resolution of nonalcoholic steatohepatitis without fibrosis worsening. Gastroenterology 150 (5), 1147–1159.e5. doi:10.1053/j.gastro.2016.01.038

PubMed Abstract | CrossRef Full Text | Google Scholar

Rigano, D., Sirignano, C., and Taglialatela-Scafati, O. (2017). The potential of natural products for targeting PPARα. Acta Pharm. Sin. B 7 (4), 427–438. doi:10.1016/j.apsb.2017.05.005

PubMed Abstract | CrossRef Full Text | Google Scholar

Romano, B., Pagano, E., Iannotti, F. A., Piscitelli, F., Brancaleone, V., Lucariello, G., et al. (2022). N-Acylethanolamine acid amidase (NAAA) is dysregulated in colorectal cancer patients and its inhibition reduces experimental cancer growth. Br. J. Pharmacol. 179 (8), 1679–1694. doi:10.1111/bph.15737

PubMed Abstract | CrossRef Full Text | Google Scholar

Sawayama, H., Ishimoto, T., Watanabe, M., Yoshida, N., Sugihara, H., Kurashige, J., et al. (2014). Small molecule agonists of PPAR-γ exert therapeutic effects in esophageal cancer. Cancer Res. 74 (2), 575–585. doi:10.1158/0008-5472.CAN-13-1836

PubMed Abstract | CrossRef Full Text | Google Scholar

Shah, M. A., Kennedy, E. B., Catenacci, D. V., Deighton, D. C., Goodman, K. A., Malhotra, N. K., et al. (2020). Treatment of locally advanced esophageal carcinoma: ASCO guideline. J. Clin. Oncol. 38 (23), 2677–2694. doi:10.1200/JCO.20.00866

PubMed Abstract | CrossRef Full Text | Google Scholar

Shi, J.-F., Wang, L., Ran, J.-C., Liu, C. C., and Zhang, H. Z. (2021). Clinical characteristics, medical service utilization, and expenditure for colorectal cancer in China, 2005 to 2014: Overall design and results from a multicenter retrospective epidemiologic survey. Cancer 127 (11), 1880–1893. doi:10.1002/cncr.33445

PubMed Abstract | CrossRef Full Text | Google Scholar

Short, M. W., Burgers, K. G., and Fry, V. T. (2017). Esophageal cancer. Am. Fam. Physician 95 (1), 22–28.

PubMed Abstract | Google Scholar

Siegel, R. L., Miller, K. D., Fuchs, H. E., and Jemal, A. (2022). Cancer statistics, 2022. CA: a Cancer J. For Clin. 72 (1), 7–33. doi:10.3322/caac.21708

CrossRef Full Text | Google Scholar

Sung, H., Ferlay, J., Siegel, R. L., Laversanne, M., Soerjomataram, I., Jemal, A., et al. (2021). Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA: a Cancer J. For Clin. 71 (3), 209–249. doi:10.3322/caac.21660

CrossRef Full Text | Google Scholar

Terashita, Y., Sasaki, H., Haruki, N., Nishiwaki, T., Ishiguro, H., Shibata, Y., et al. (2002). Decreased peroxisome proliferator-activated receptor gamma gene expression is correlated with poor prognosis in patients with esophageal cancer. Jpn. J. Clin. Oncol. 32 (7), 238–243. doi:10.1093/jjco/hyf056

PubMed Abstract | CrossRef Full Text | Google Scholar

Terzić, J., Grivennikov, S., Karin, E., and Karin, M. (2010). Inflammation and colon cancer. Gastroenterology 138 (6), 2101–2114. doi:10.1053/j.gastro.2010.01.058

PubMed Abstract | CrossRef Full Text | Google Scholar

van den Brandt, P. A. (2022). The impact of a healthy lifestyle on the risk of esophageal and gastric cancer subtypes. Eur. J. Epidemiol. 37 (9), 931–945. doi:10.1007/s10654-022-00899-w

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, R., Li, J., Zhou, X., Mao, Y., Wang, W., Gao, S., et al. (2022). Single-cell genomic and transcriptomic landscapes of primary and metastatic colorectal cancer tumors. Genome Med. 14 (1), 93. doi:10.1186/s13073-022-01093-z

PubMed Abstract | CrossRef Full Text | Google Scholar

Wu, H., Liu, B., Chen, Z., Li, G., and Zhang, Z. (2020). MSC-induced lncRNA HCP5 drove fatty acid oxidation through miR-3619-5p/AMPK/PGC1α/CEBPB axis to promote stemness and chemo-resistance of gastric cancer. Cell Death Dis. 11 (4), 233. doi:10.1038/s41419-020-2426-z

PubMed Abstract | CrossRef Full Text | Google Scholar

Wu, K., Yang, Y., Liu, D., Qi, Y., Zhang, C., Zhao, J., et al. (2016). Activation of PPARγ suppresses proliferation and induces apoptosis of esophageal cancer cells by inhibiting TLR4-dependent MAPK pathway. Oncotarget 7 (28), 44572–44582. doi:10.18632/oncotarget.10067

PubMed Abstract | CrossRef Full Text | Google Scholar

Xiao, L., and Wang, N. (2022). PPAR-δ: A key nuclear receptor in vascular function and remodeling. J. Mol. Cell. Cardiol. 169, 1–9. doi:10.1016/j.yjmcc.2022.04.019

PubMed Abstract | CrossRef Full Text | Google Scholar

Xiao, X.-J., and Zheng, H.-C. (2020). NDRG1 was downregulated and worked as favorable biomarker in the development of gastric cancer. Transl. Cancer Res. 9 (1), 210–221. doi:10.21037/tcr.2019.12.76

PubMed Abstract | CrossRef Full Text | Google Scholar

Yamamoto, K., Tamura, T., Henmi, K., Kuboyama, T., Yanagisawa, A., Matsubara, M., et al. (2018). Development of dihydrodibenzooxepine peroxisome proliferator-activated receptor (PPAR) gamma ligands of a novel binding mode as anticancer agents: Effective mimicry of chiral structures by olefinic E/Z-isomers. J. Med. Chem. 61 (22), 10067–10083. doi:10.1021/acs.jmedchem.8b01200

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, X., Zhang, L., Lai, J., Lu, Y., Ye, J., and Wang, Y. (2022). Screening and identification of hub gene and differential gene and mutation sequence analysis of related genes in colorectal cancer based on bioinformatics analysis. J. Gastrointest. Oncol. 13 (6), 3056–3066. doi:10.21037/jgo-22-1131

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhao, D., Ma, Y., Li, X., and Lu, X. (2019). microRNA-211 promotes invasion and migration of colorectal cancer cells by targeting FABP4 via PPARγ. J. Cell. Physiology 234 (9), 15429–15437. doi:10.1002/jcp.28190

CrossRef Full Text | Google Scholar

Zhou, J., Yang, L., Li, Y., Arbman, G., Chen, K. L., Zhou, B., et al. (2014). The prognostic significance of peroxisome proliferator-activated receptor β expression in the vascular endothelial cells of colorectal cancer. J. Gastroenterology 49 (3), 436–445. doi:10.1007/s00535-013-0845-7

CrossRef Full Text | Google Scholar

Keywords: PPARs, esophageal cancer, cancer progression, gastric cancer, colorectal cancer, cancer treatment

Citation: Zhang M and He S (2023) Peroxisome proliferator-activated receptors regulate the progression and treatment of gastrointestinal cancers. Front. Pharmacol. 14:1169566. doi: 10.3389/fphar.2023.1169566

Received: 19 February 2023; Accepted: 06 March 2023;
Published: 21 March 2023.

Edited by:

Yingkun Xu, Chongqing Medical University, China

Reviewed by:

Qian Xiang, Peking University Third Hospital, China
Shishuo Sun, Xuzhou Medical University, China
Zhixian Chen, The University of Hong Kong, Hong Kong SAR, China

Copyright © 2023 Zhang and He. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Shujie He, shujiehe@foxmail.com

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.