Therapeutic Potential of Peroxisome Proliferator-Activated Receptors in Chronic Inflammation and Colorectal Cancer




Peroxisome proliferatoreactivated receptors (PPARs) are members of the nuclear hormone receptor superfamily and have been implicated in a variety of physiologic and pathologic processes, such as nutrient metabolism, energy homeostasis, inflammation, and cancer. This article highlights breakthroughs in our understanding of the potential roles of PPARs in inflammatory bowel disease and colorectal cancer. PPARs might hold the key to some of the questions that are pertinent to the pathophysiology of inflammatory diseases and colorectal cancer and could possibly serve as drug targets for new antiinflammatory therapeutic and anticancer agents.


The recognition of chronic inflammation caused by infections or autoimmune diseases as the seventh trait of cancer has highlighted the contribution of inflamed stroma to tumor initiation, growth, and metastasis. Epidemiologic studies indicate that chronic inflammation is clearly associated with increased risk of cancers in several instances, including esophageal, gastric, hepatic, pancreatic, and colorectal cancers (CRC). For example, it has been long known that patients with persistent hepatitis B infection, Helicobacter pylori infection, or an immune disorder, such as inflammatory bowel disease (IBD), have a higher risk for developing liver or gastrointestinal tract cancer. It has been estimated that chronic inflammation contributes to the development of approximately 15% of malignancies worldwide. The best evidence for the link between inflammation and tumor progression comes from recent epidemiologic studies and clinical trials that have shown that long-term use of nonsteroidal antiinflammatory drugs (NSAIDs) reduce the relative risk of developing CRC by 40% to 50%.


The gastrointestinal mucosa forms a complex semipermeable barrier between the host and the largest source of foreign antigens. The mucosal barrier consists of epithelial cell junctions and the underlying stromal elements including immune cells. An abnormal mucosal immune response to bacteria, which make up the intestinal flora, is thought to result in chronic inflammation and the development of IBD. IBD, with its two clinical manifestations of Crohn disease (CD) and ulcerative colitis (UC), is a chronic inflammatory disorder of the gastrointestinal tract. Chronic IBD (especially pancolitis) significantly increases the risk of developing CRC. The observation that 5-aminosalicylic acid (5-ASA), currently used in the treatment of UC, suppresses the development of colitis-associated cancer in an animal model supports this notion.


A large body of evidence indicates that genetic mutations, epigenetic changes, chronic inflammation, diet, and lifestyle are risk factors for cancer. Similar to other solid tumors, CRC is a heterogeneous disease with at least 3 major forms: hereditary, sporadic, and colitis-associated CRCs. Patients with familial adenomatous polyposis (FAP), caused by a germline mutation in 1 allele of the tumor suppressor gene adenomatous polyposis coli ( APC ), have a near 100% risk of developing CRC by the age of 40 years, if untreated. Somatic loss of APC function occurs in about 85% of sporadic colorectal adenomas and carcinomas. Hereditary nonpolyposis CRC, which is caused by inherited mutations in genes for DNA mismatch repair, such as MLH1 , MSH2 , and MSH6 , is responsible for approximately 2% to 7% of all diagnosed cases of CRC. The average age at which patients with this syndrome develop cancer is around 44 years as compared with 64 years in the general population. Together with FAP and hereditary nonpolyposis CRC, IBD is among the top 3 high-risk conditions for CRC; therefore, patients with IBD face an increased lifetime risk for developing CRC. Compared with sporadic CRC, colitis-associated CRC affects individuals at a younger age.


Peroxisome proliferator–activated receptors (PPARs), which were initially identified as mediators of the peroxisome proliferators in the early 1990s, belong to the nuclear hormone receptor superfamily and are also ligand-dependent transcription factors. PPARs play a central role in regulating the storage and catabolism of dietary fats via complex metabolic pathways, including fatty acid oxidation and lipogenesis. To date, 3 mammalian PPARs have been identified and are referred to as PPARα (NR1C1), PPARδ/β (NR1C2), and PPARγ (NR1C3). Each PPAR isotype displays a tissue-selective expression pattern. PPARα and PPARγ are predominantly present in the liver and adipose tissue, respectively, whereas PPARδ is expressed in diverse tissues, and its expression in the gastrointestinal tract is very high compared with that in other tissues. As ligand-dependent transcription factors, transcriptional activation by PPARs depends on ligand binding and the interaction of coregulators. PPAR ligands are chemically unrelated molecules, including a variety of fatty acids, fatty acid derivatives, and steroids as well as synthetic compounds. Polyunsaturated fatty acids activate all 3 PPAR isotypes with relatively low affinity. The endogenous fatty acid derivatives, which are mainly converted by cyclooxygenase (COX) and lipoxygenase enzymes, selectively bind and activate each PPAR isotype. For example, 15-deoxy-Δ 1214 prostaglandin (PG) J 2 (15dPGJ 2 ), a dehydration product of PGD 2 , is a natural ligand for PPARγ, whereas PGI 2 can transactivate PPARδ.


It is well established that modulation of PPAR activity maintains cellular and whole-body glucose and lipid homeostasis. Hence, great efforts have been made to develop drugs targeting these receptors. For example, PPARγ synthetic agonists, such as troglitazone, rosiglitazone, and pioglitazone, are clinically used for the therapy for non–insulin-dependent diabetes mellitus. The antiatherosclerotic and hypolipidemic agents including fenofibrate and gemfibrozil are PPARα synthetic agonists that induce hepatic lipid uptake and catabolism. Genetic and pharmacologic studies have also revealed that PPARδ agonists are potential drugs for use in the treatment of dyslipidemias, obesity, and insulin resistance. Therefore, at present, the PPARδ agonist (GW501516) is in phase 3 clinical trials for the evaluation of the treatment of patients with hyperlipidemias and obesity. In addition to modulation of lipid homeostasis and energy balance, PPARs have emerged as essential molecules in the pathogenesis of IBD and CRC.


PPARs and IBD


The currently available therapies for IBD include 5-ASA, corticosteroids, antibiotics, immune modulators, and immunosuppressive agents such as azathioprine, 6-mercaptopurine, and cyclosporine. Corticosteroids and immunosuppressive agents are associated with significant risks of unwanted side effects, and not all patients respond to these medications. 5-ASA agents are generally safe but induce remission only in approximately 50% of patients with UC. It is, therefore, essential to develop newer therapeutic interventions for patients with IBD. A growing body of evidence indicates that PPARα and PPARγ have an antiinflammatory effect on IBD, and the agonists of these 2 receptors might serve as a new class of effective therapeutic agents for IBD. The role of PPARδ in IBD remains ambiguous and deserves significant attention, and future research must be directed to better understand the role of PPARs in regulating chronic inflammation in IBD.


PPARα


PPARα is highly expressed in mouse colonic epithelial cells facing the intestinal lumen, and its expression is induced by glucocorticoids (GCs). Subsequent studies have further demonstrated that PPARα mediates the antiinflammatory effects of GC in a mouse model of chemically induced colitis. In this study, treatment with dexamethasone, a potent synthetic member of the GC class of steroid drugs, suppressed the formation of dinitrobenzene sulfuric acid (DNBS)–induced colitis in wild-type mice but not in PPARα knockout mice. Consistent with these results, the deletion of PPARα promoted the severity of colitis in DNBS-treated mice, whereas activation of PPARα by its agonist significantly reduced colonic inflammation in this mouse model. However, there is no report thus far on the precise role of PPARα in genetic models of IBD (transgenic and knockout models).


PPARγ


Although PPARγ is predominantly present in the liver and adipose tissue, it is also expressed in the intestinal epithelium, immune cells, and adipocytes. However, patients with UC, but not those with CD, show decreased PPARγ levels in colonic epithelial cells compared with normal controls. This observation raises the hypothesis that microbe-host interactions, chronic inflammation, and/or genetic predisposition may lead to low PPARγ levels in colonic epithelial cells, which in turn may result in unrestrained inflammation. Several lines of evidence support the notion that PPARγ may serve as a new therapeutic target in IBD. In mouse models of chemically induced colitis, 5-ASA treatment had a beneficial effect on colitis only in wild-type mice and not in heterozygous PPARγ +/− mice, demonstrating that PPARγ mediates the antiinflammatory effect of 5-ASA. Furthermore, treatment with a PPARγ ligand, thiazolidinedione, markedly reduced colonic inflammation in mouse models of chemically induced colitis and interleukin (IL)-10 deficient mice (a genetic model of colitis), suggesting that activation of PPARγ suppresses inflammation in IBD.


Because PPARγ is expressed in intestinal epithelial cells, macrophages, and T and B lymphocytes, it is critical to understand the contribution of PPARγ present in each of these cell types to this protection. The results from 2 studies showed that the disruption of PPARγ in colonic epithelial cells worsened colonic inflammatory lesions in dextran sulfate sodium (DSS)–treated mice, indicating that PPARγ expression in epithelial cells is required for the prevention of experimental IBD. Similarly, mice with deficiency of PPARγ in CD4 T lymphocytes are more sensitive to trinitrobenzene sulfonic acid–induced colitis because the deficiency of PPARγ in regulatory T cells impaired their ability to prevent effector CD4 T lymphocyte–induced colitis. Moreover, mice with a targeted disruption of PPARγ in macrophages displayed an increased susceptibility to DSS-induced colitis compared with wild-type littermates, demonstrating that PPARγ is required for macrophage-mediated protection against colitis. Consistent with these results, an increase in PPARγ expression by adenovirus-mediated gene transfer attenuated colonic inflammation induced by DSS in mice. In addition, a recent study showed that the antiinflammatory effects of PPARγ on IBD are via maintenance of innate antimicrobial immunity in the mouse colon. The results from 1 randomized placebo-controlled trial and 1 open-label trial showed that a PPARγ agonist, rosiglitazone, has therapeutic efficacy in humans with UC. Collectively, all of these studies support a rationale to develop PPARγ agonists as potential therapeutic and prophylactic agents against IBD.


PPARδ


Little is known about the role of PPARδ in IBD, and the results from 2 mouse models of IBD are controversial. Deletion of PPARδ significantly exacerbated colitis, whereas treatment with a PPARδ agonist did not affect the clinical symptoms in the DSS-treated mouse model. This study implies that PPARδ, like PPARγ, exerts antiinflammatory effects in IBD via a ligand-independent mechanism. In contrast with this observation, administration of a PPARδ agonist caused enhanced colitis in IL-10–deficient mice (a genetic model of colitis), suggesting that PPARδ has a proinflammatory effect. Therefore, further studies are necessary to clarify the biologic functions of PPARδ in the modulation of IBD.




PPARs and CRC


In addition to these metabolic and inflammatory properties, the roles of PPARs in CRC progression have been extensively investigated. PPARs can function as either tumor suppressors or accelerators, suggesting that these receptors are potential candidates as drug targets for cancer prevention and treatment.


PPARα


Less is known about the role of PPARα in human cancer, although long-term administration of a PPARα agonist induces the development of hepatocarcinomas in mice but not in PPARα-null animals, conclusively demonstrating that PPARα mediates these effects in promoting liver cancer. In spite of the fact that activation of PPARα by exogenous agonists generally causes inhibition of tumor cell growth in cell lines derived from CRC, melanoma, and glial brain tumors, the physiologic significance of PPARα in the regulation of CRC progression is also less well characterized than that of PPARγ and PPARδ.


PPARγ


Because of the elevated expression of PPARγ in CRC and its involvement in regulating cellular differentiation, PPARγ has become a point of interest in CRC studies. However, studies of PPARγ mutation in human colon tumor samples and CRC cell lines have produced controversial results. One study has shown that 8% of the patients with primary human CRCs had a loss-of-function mutation in 1 allele of the PPARγ gene. Recent data revealed that a Pro12Ala polymorphism in the PPARγ gene is associated with an increased risk of CRC. These results suggest a putative role for this receptor as a tumor suppressor. In contrast, another study showed that the mutant PPARγ gene was not detected in human colon carcinoma samples or CRC cell lines, suggesting that PPARγ mutations in human CRC may be a rare event.


It is well established in numerous in vitro studies that activation of PPARγ results in growth arrest of colon carcinoma cells through induction of cell cycle arrest or/and apoptosis. However, the effect of PPARγ on CRC progression in vivo is controversial because of conflicting results from different mouse models of colon cancer. Although PPARγ agonists inhibit colorectal carcinogenesis in xenograft models and azoxymethane (AOM)-induced colon cancer model, these drugs are reported to have either tumor-promoting or tumor-inhibiting effects in a mouse model of FAP, the Apc Min/+ mouse. Multiple studies showed that administration of PPARγ agonists significantly increases the number of colon adenomas in the Apc Min/+ and even in wild-type C57BL/6 mice. However, other studies showed that treatment of 2 different Apc mutant models ( Apc Min/+ and Apc Δ1309 ) with a PPARγ agonist pioglitazone reduced the polyp number in small and large intestines in a dose-dependent manner. These divergent effects of PPARγ might be related to drug doses and bioavailability and/or the animal models that are used. These paradoxic observations seem to have been resolved by genetic studies, showing that the heterozygous disruption of PPARγ is sufficient to increase tumor numbers in AOM-treated mice and that intestinal-specific PPARγ knockout promotes tumor growth in Apc Min/+ mice. Thus, genetic evidence supports the hypothesis that PPARγ serves as a tumor suppressor in CRC. In addition, a combined treatment of mice with a selective COX-2 inhibitor and a PPARγ agonist significantly inhibited the incidence and multiplicity of inflammation-associated colonic adenocarcinoma induced by AOM or DSS. A retrospective cohort study revealed that treatment of diabetic patients with a PPARγ agonist (thiazolidinedione) exhibited a mild trend toward risk reduction of CRC, although this difference did not reach statistical significance. Collectively, these findings further support the rationale of developing PPARγ agonists as antitumor agents.


PPARδ


The role of PPARδ in colorectal carcinogenesis is more controversial than that of PPARγ. The first evidence linking PPARδ to carcinogenesis actually came from studies on gastrointestinal cancer. PPARδ was identified as a direct transcriptional target of APC/β-catenin/Tcf pathway and as a repression target of NSAIDs. A large case-control study showed that the protective effect of NSAIDs against colorectal adenomas was modulated by a polymorphism in the PPAR gene. Moreover, COX-2-derived PGI 2 directly transactivates PPARδ, and COX-2-derived PGE 2 indirectly induces PPARδ activation in CRC, hepatocellular carcinoma, and cholangiocarcinoma cells. In addition, PPARδ expression and activity are also induced by oncogenic K-Ras. These studies indicate that PPARδ is a focal point of crosstalk between oncogenic signaling pathways.


Similar to PPARγ, investigation of PPARδ expression in human and mouse colonic tumor samples and CRC cell lines generated controversial results. Some reports showed that PPARδ is elevated in most human CRCs and in tumors arising in the Apc Min/+ mice and AOM-treated rats, in agreement with the observations that activation of the β-catenin/Tcf pathway by APC mutation or K-Ras upregulates PPARδ expression. PPARδ proteins are accumulated only in human CRC cells with highly malignant morphology. Downregulation of PPARδ is correlated with antitumor effects of dietary fish oil and pectin in rats treated with radiation and AOM. However, other reports showed that PPARδ expression is lower in human cancer tissues and adenomas from the Apc Min/+ mice than in normal control tissues.


In a murine xenograft cancer model, the disruption of both PPARδ alleles by deletion of its exons 4 and 6 in human HCT116 colon carcinoma cells decreased tumorigenicity, suggesting that activation of PPARδ promotes tumor growth. To further determine whether PPARδ attenuates or promotes intestinal tumor growth, 3 mouse models of CRC were used, including AOM-treated, Apc Min/+ , and Mlh -null mice. Mlh is a DNA mismatch repair gene that is involved in hereditary nonpolyposis CRC. Conflicting data were obtained from studies in AOM-treated and Apc Min/+ mice. For example, activation of PPARδ by a selective synthetic PPARδ agonist (GW501516) or a PPARδ endogenous activator (PGE 2 ) accelerated intestinal adenoma growth in Apc Min/+ mice by promoting tumor cell survival. In contrast, another PPARδ ligand (GW0742) inhibited colon carcinogenesis in AOM-treated mice but promoted small intestinal polyp growth in Apc Min/+ mice. It is not clear whether PPARδ mediates the effects of GW0742 in Apc Min/+ mice in this study. A genetic study showed that loss of PPARδ by deletion of its exons 4 and 5 attenuated small and large intestinal adenoma growth and demonstrated that PPARδ mediated the tumor-promoting effects of the PPARδ ligand (GW501516) and PGE 2 in Apc Min/+ mice. In a recent study with a tissue-specific deletion of PPARδ exon 4 in the colon, loss of PPARd inhibited colonic carcinogenesis in AOM-treated mice, further confirming the notion that PPARδ serves as a tumor accelerator. On the other hand, several other studies have shown different results when using PPARδ mutant mice generated by germline deletion of PPARδ exon 8. Deletion of PPARδ exon 8 enhances polyp growth in Apc Min/+ and AOM-treated mice in the absence of exogenous PPARδ stimulation. In Mlh -null mice, no significant differences are evident in the number and size of intestinal adenomas between wild-type and PPARδ mutant mice (deletion of PPARδ exon 8). The conflicting results regarding the effect of PPARδ on intestinal tumorigenesis in Apc Min/+ and AOM-treated mice could be attributed to differences in the specific targeting strategy used to delete PPARδ. Deletion of PPARδ exon 4 and/or 5, which encode an essential portion of the DNA binding domain, is thought to disrupt PPARδ’s function as a nuclear transcription factor and inhibit tumorigenesis. The deletion of exon 8, the last PPARδ exon, is postulated to generate a hypomorphic allele, which retains some aporeceptor function. Indeed, the observation that the high rates of embryonic mortality and abnormal placental development occurred in mice in which PPARδ exons 4 and 5 were deleted but not in mice in which PPARδ exon 8 was deleted supports this hypothesis. Taken together, not enough evidence is available to establish whether PPARδ has pro- or antitumorigenic effect on CRC progression, and the role of PPARδ in cancer biology remains unclear.




PPARs and CRC


In addition to these metabolic and inflammatory properties, the roles of PPARs in CRC progression have been extensively investigated. PPARs can function as either tumor suppressors or accelerators, suggesting that these receptors are potential candidates as drug targets for cancer prevention and treatment.


PPARα


Less is known about the role of PPARα in human cancer, although long-term administration of a PPARα agonist induces the development of hepatocarcinomas in mice but not in PPARα-null animals, conclusively demonstrating that PPARα mediates these effects in promoting liver cancer. In spite of the fact that activation of PPARα by exogenous agonists generally causes inhibition of tumor cell growth in cell lines derived from CRC, melanoma, and glial brain tumors, the physiologic significance of PPARα in the regulation of CRC progression is also less well characterized than that of PPARγ and PPARδ.


PPARγ


Because of the elevated expression of PPARγ in CRC and its involvement in regulating cellular differentiation, PPARγ has become a point of interest in CRC studies. However, studies of PPARγ mutation in human colon tumor samples and CRC cell lines have produced controversial results. One study has shown that 8% of the patients with primary human CRCs had a loss-of-function mutation in 1 allele of the PPARγ gene. Recent data revealed that a Pro12Ala polymorphism in the PPARγ gene is associated with an increased risk of CRC. These results suggest a putative role for this receptor as a tumor suppressor. In contrast, another study showed that the mutant PPARγ gene was not detected in human colon carcinoma samples or CRC cell lines, suggesting that PPARγ mutations in human CRC may be a rare event.


It is well established in numerous in vitro studies that activation of PPARγ results in growth arrest of colon carcinoma cells through induction of cell cycle arrest or/and apoptosis. However, the effect of PPARγ on CRC progression in vivo is controversial because of conflicting results from different mouse models of colon cancer. Although PPARγ agonists inhibit colorectal carcinogenesis in xenograft models and azoxymethane (AOM)-induced colon cancer model, these drugs are reported to have either tumor-promoting or tumor-inhibiting effects in a mouse model of FAP, the Apc Min/+ mouse. Multiple studies showed that administration of PPARγ agonists significantly increases the number of colon adenomas in the Apc Min/+ and even in wild-type C57BL/6 mice. However, other studies showed that treatment of 2 different Apc mutant models ( Apc Min/+ and Apc Δ1309 ) with a PPARγ agonist pioglitazone reduced the polyp number in small and large intestines in a dose-dependent manner. These divergent effects of PPARγ might be related to drug doses and bioavailability and/or the animal models that are used. These paradoxic observations seem to have been resolved by genetic studies, showing that the heterozygous disruption of PPARγ is sufficient to increase tumor numbers in AOM-treated mice and that intestinal-specific PPARγ knockout promotes tumor growth in Apc Min/+ mice. Thus, genetic evidence supports the hypothesis that PPARγ serves as a tumor suppressor in CRC. In addition, a combined treatment of mice with a selective COX-2 inhibitor and a PPARγ agonist significantly inhibited the incidence and multiplicity of inflammation-associated colonic adenocarcinoma induced by AOM or DSS. A retrospective cohort study revealed that treatment of diabetic patients with a PPARγ agonist (thiazolidinedione) exhibited a mild trend toward risk reduction of CRC, although this difference did not reach statistical significance. Collectively, these findings further support the rationale of developing PPARγ agonists as antitumor agents.


PPARδ


The role of PPARδ in colorectal carcinogenesis is more controversial than that of PPARγ. The first evidence linking PPARδ to carcinogenesis actually came from studies on gastrointestinal cancer. PPARδ was identified as a direct transcriptional target of APC/β-catenin/Tcf pathway and as a repression target of NSAIDs. A large case-control study showed that the protective effect of NSAIDs against colorectal adenomas was modulated by a polymorphism in the PPAR gene. Moreover, COX-2-derived PGI 2 directly transactivates PPARδ, and COX-2-derived PGE 2 indirectly induces PPARδ activation in CRC, hepatocellular carcinoma, and cholangiocarcinoma cells. In addition, PPARδ expression and activity are also induced by oncogenic K-Ras. These studies indicate that PPARδ is a focal point of crosstalk between oncogenic signaling pathways.


Similar to PPARγ, investigation of PPARδ expression in human and mouse colonic tumor samples and CRC cell lines generated controversial results. Some reports showed that PPARδ is elevated in most human CRCs and in tumors arising in the Apc Min/+ mice and AOM-treated rats, in agreement with the observations that activation of the β-catenin/Tcf pathway by APC mutation or K-Ras upregulates PPARδ expression. PPARδ proteins are accumulated only in human CRC cells with highly malignant morphology. Downregulation of PPARδ is correlated with antitumor effects of dietary fish oil and pectin in rats treated with radiation and AOM. However, other reports showed that PPARδ expression is lower in human cancer tissues and adenomas from the Apc Min/+ mice than in normal control tissues.


In a murine xenograft cancer model, the disruption of both PPARδ alleles by deletion of its exons 4 and 6 in human HCT116 colon carcinoma cells decreased tumorigenicity, suggesting that activation of PPARδ promotes tumor growth. To further determine whether PPARδ attenuates or promotes intestinal tumor growth, 3 mouse models of CRC were used, including AOM-treated, Apc Min/+ , and Mlh -null mice. Mlh is a DNA mismatch repair gene that is involved in hereditary nonpolyposis CRC. Conflicting data were obtained from studies in AOM-treated and Apc Min/+ mice. For example, activation of PPARδ by a selective synthetic PPARδ agonist (GW501516) or a PPARδ endogenous activator (PGE 2 ) accelerated intestinal adenoma growth in Apc Min/+ mice by promoting tumor cell survival. In contrast, another PPARδ ligand (GW0742) inhibited colon carcinogenesis in AOM-treated mice but promoted small intestinal polyp growth in Apc Min/+ mice. It is not clear whether PPARδ mediates the effects of GW0742 in Apc Min/+ mice in this study. A genetic study showed that loss of PPARδ by deletion of its exons 4 and 5 attenuated small and large intestinal adenoma growth and demonstrated that PPARδ mediated the tumor-promoting effects of the PPARδ ligand (GW501516) and PGE 2 in Apc Min/+ mice. In a recent study with a tissue-specific deletion of PPARδ exon 4 in the colon, loss of PPARd inhibited colonic carcinogenesis in AOM-treated mice, further confirming the notion that PPARδ serves as a tumor accelerator. On the other hand, several other studies have shown different results when using PPARδ mutant mice generated by germline deletion of PPARδ exon 8. Deletion of PPARδ exon 8 enhances polyp growth in Apc Min/+ and AOM-treated mice in the absence of exogenous PPARδ stimulation. In Mlh -null mice, no significant differences are evident in the number and size of intestinal adenomas between wild-type and PPARδ mutant mice (deletion of PPARδ exon 8). The conflicting results regarding the effect of PPARδ on intestinal tumorigenesis in Apc Min/+ and AOM-treated mice could be attributed to differences in the specific targeting strategy used to delete PPARδ. Deletion of PPARδ exon 4 and/or 5, which encode an essential portion of the DNA binding domain, is thought to disrupt PPARδ’s function as a nuclear transcription factor and inhibit tumorigenesis. The deletion of exon 8, the last PPARδ exon, is postulated to generate a hypomorphic allele, which retains some aporeceptor function. Indeed, the observation that the high rates of embryonic mortality and abnormal placental development occurred in mice in which PPARδ exons 4 and 5 were deleted but not in mice in which PPARδ exon 8 was deleted supports this hypothesis. Taken together, not enough evidence is available to establish whether PPARδ has pro- or antitumorigenic effect on CRC progression, and the role of PPARδ in cancer biology remains unclear.

Only gold members can continue reading. Log In or Register to continue

Stay updated, free articles. Join our Telegram channel

Feb 26, 2017 | Posted by in GASTROENTEROLOGY | Comments Off on Therapeutic Potential of Peroxisome Proliferator-Activated Receptors in Chronic Inflammation and Colorectal Cancer

Full access? Get Clinical Tree

Get Clinical Tree app for offline access