Lab Notes

Peering into the Future: The Game-Changing Benefits of PPAR agonists in the skin

Unleash the potential of peroxisome proliferator-activated receptors (PPARs) for optimal skin health! These transcription factors play a critical role in regulating various cellular processes, including inflammation, differentiation, and proliferation, in skin cells. By modulating these pathways, PPARs can exert a significant impact on the skin's physiology and function, offering a plethora of benefits for skin barrier health. Discover the latest scientific findings on the mechanisms of PPARs in skin and explore the cutting-edge skincare ingredients that leverage their therapeutic potential. 

An Intro to Nuclear Hormone Receptor

I’m sure you are well familiar with Retinoids by now. Biochemically speaking, Retinoids are a group of molecules that can bind and activate the Nuclear Hormone Receptor, Retinoid Acid Receptors (RAR) and Retinoid X Receptors (RXR). Nuclear Hormone Receptors are present in the nuclear envelope, and once activated, they translocate inside the nucleus. They then dimerize with other Nuclear Hormone Receptors to activate, repress or modify DNA expression. This ability to modify DNA expression is why Retinoids have multiple benefits for the skin compared to actives that only work on the protein level.

What are PPARs?

PPAR (Peroxisome Proliferator-Activated Receptor) is another group of Nuclear Hormone Receptors similar to RAR and RXR. PPAR agonists are molecules that can bind and activate PPARs, just like Retinoids bind RARs. They contain four domains: 

There are three common types in the skin (1):

Each of these isotypes has its function in the skin, but collectively, we call them the master orchestrators of barrier function and inflammation. 

What are PPARs’ functions in the skin?

PPARs have a variety of functions in the skin, and as mentioned above, most are related to barrier function and inflammation:

Barrier Lipid Production

You know by now that the significant part of the moisture barrier is the barrier lipids (Ceramides, Cholesterol and Fatty Acids). In a normal state, the skin produces fatty acids from the basal layer to the Stratum Granulosum. This increase in fatty acid content in the cell layers activates PPARs, as fatty acids are one of the most common PPAR agonists. PPAR activation leads to the enhancement of Cholesterol and Ceramide production. (2) It is a feed-forward system where the resulting Fatty acid, Ceramide and Cholesterol can activate further PPAR resulting in an abundance of these Barrier lipids in the Stratum Granulosum. In essence, PPAR here acts as an orchestrator to ensure enough of each barrier lipid in the skin for lamellar body production.

Adapted from: Feingold, K. R. (2007). Thematic review series: skin lipids. The role of epidermal lipids in cutaneous permeability barrier homeostasis. Journal of Lipid Research, 48(12), 2531–2546.

Lamellar Body Formation and Secretion

For barrier lipids to be secreted, they must first be packed into sacs called lamellar bodies. Fatty acids serve two purposes here: 1) serve as a building block for Ceramides and 2) convert them into Phospholipids to be packed into lamellar bodies. On the other hand, Ceramides need to be converted into Glucosylceramide (the more water-soluble ceramide) and Sphingomyelin. Finally, Cholesterol is just packaged up as Cholesterol in lamellar bodies.

The role of PPAR here is during the transport and packaging of lamellar bodies. PPAR seems to initiate the lamellar body formation through the action of ATP-Binding Cassette Transporter family: ABCG1 and ABCA12. ABCG1 forms the lamellar membrane and recruits the lamellar lipids to be packaged (3), while ABCA12 transports Glucosylceramide into the lamellar bodies. (4) Once the lamellar bodies are formed, the increase in PPAR expression due to barrier lipid formation causes the secretion of lamellar bodies in the Stratum Granulosum (SG)-Stratum Corneum(SC) interface. (5)

Processing of Precursor Lipids

Once the lamellar bodies are secreted in the SG-SC interface, these barrier lipids are not yet fully processed. For them to properly merge with the existing lamellar layer, the Glucosylceramides and Sphingomyelin need to be processed into Ceramides, and the Phospholipids need to be converted into Fatty acids. Incidentally, the processing enzymes (inactive form) have also been packed in the lamellar bodies. Once they reach the SC, these processing enzymes activate and start the conversion of the precursor lipids. One enzyme in particular (B-glucocerebrosidase) is essential in forming functional ceramides. It is active around a pH of 5, and its activity is increased by PPAR activation. (6)

Corneocyte Maturation

Aside from the barrier lipids, the other major moisture barrier component is the corneocyte (aka the bricks). As the Keratinocytes differentiate, their membrane gets bolstered with a rigid envelope composed of Involucrin, Loricrin and other proteins. This cornified envelope becomes more compact and makes the corneocyte more resistant to chemical and mechanical damage. In addition, the enzyme Transglutaminase-1 attaches certain Ceramides to the cell, coating the cell in a lipid envelope. This lipid envelope serves as an attachment point for lamellar layers.

PPAR’s role here is to push forward cell differentiation. It is interesting to note that PPAR appears to be the main orchestrating body coordinating barrier lipid formation, lamellar body secretion and cornification in cell differentiation.

Antimicrobial Peptides

In addition to the barrier lipids and processing enzymes in lamellar bodies, some short-peptides called Antimicrobial Peptides (AMPs) are included. These peptides are one of the ways that the skin fights off microbes, as they are effective against various Bacteria, Fungi and Viruses. (Side note: In Rosacea, there is a decrease in AMPs in the skin, which leads to easier colonization by harmful bacteria or fungi). One particular AMP is β-Defensin. PPAR activation also significantly increases βD2 and βD3 in the skin contributing to the protective function of the newly formed barrier. (7)

Wound Healing

PPAR’s function in wound healing is quite interesting. When a wound is present in the skin, there are multiple inflammatory responses within the wound. One of these inflammatory responses is TNFa-induced apoptosis which essentially kills all the affected cells in the wound. PPAR expression was seen to be increased in the wound edge in non-affected cells. Its function is to confer resistance to apoptotic signals in the wound edge, creating a barrier around the wound. Once the inflammation is toned down, these barrier cells migrate and undergo differentiation. PPAR essentially organizes gene expression around the wound, leading to survival, migration and differentiation of keratinocytes. (8)

Anti-Inflammation

Inflammation is a very complex process that has a lot of triggers. Once started, it forms a cascade of pro-inflammatory molecules and pathways that activates multiple inflammatory pathways. However, as soon as the inflammation starts, the skin produces anti-inflammatory molecules. The visible inflammation in the skin results from the balance between pro-inflammatory and anti-inflammatory systems.
Most of the pro-inflammatory genes are under the control of the AP-1 and NF-kB signaling pathways. Most skin damages pass through either or both of these signaling pathways. Interestingly, PPAR was shown to tone down both NF-kB and AP-1 signaling inside the nucleus. (9) PPARa can inhibit NF-kB in multiple steps, but its action is mainly to prevent the degradation of IkBa, which is an inhibitor for NF-kB. IkBa binds to inactive NF-kB, so if there are more IkBa in the nucleus, NF-kB remains bound and won't be able to activate target genes. On the other hand, PPAR directly inhibits AP-1 signaling through the binding of c-Jun. Hence AP-1 is also toned down when PPAR is activated. PPAR also acts on two minor inflammatory signaling pathway: NFAT and STAT.

Adapted from: Delerive, P., Fruchart, J. C., & Staels, B. (2001). Peroxisome proliferator-activated receptors in inflammation control. The Journal of endocrinology, 169(3), 453–459. https://doi.org/10.1677/joe.0.1690453

What are the benefits of the topical application of PPAR agonists?

Barrier Function

Topical treatment with PPARa, PPARb/d activators increased Cholesterol, fatty acids, and sphingolipid synthesis in the epidermis. There was also an increase in the activity of Serine Palmitoyltransferase, Ceramide Synthase, and HMG-CoA synthase in human organotypic cultures with PPARa activator. On the other hand, PPARg did not exert significant effects on epidermal lipid synthesis. (10) In addition, topical treatment of adult mice with PPARa activators (Clofibrate and Farnesol) decreased epidermal thickness, while the expression of structural proteins. Although PPARa activation leads to no change in TEWL under basal conditions, it accelerated permeability barrier repair following abrogation by either tape stripping or 10% SDS treatment. (11) It is crucial to note that PPARs work on accelerating barrier repair when you have a damaged barrier. If your skin is not damaged, they don't change your basal TEWL.

Source: Elias, P. (2022). Optimizing emollient therapy for skin barrier repair in atopic dermatitis. Annals Of Allergy, Asthma &Amp; Immunology, 128(5), 505-511. https://doi.org/10.1016/j.anai.2022.01.012

Anti-inflammation

Most of the work regarding anti-inflammation is through novel PPAR compounds targeted toward inflammatory diseases. Examples of these are GED-0507-34L (PPARg agonist), GW7647 (PPARa), and Wy-14643 (PPARa). There are some PPARg ligand studies, but the anti-inflammatory effect is mainly through PPARa. Natural ligands like Palmitoylethanolamide were also studied for their anti-inflammatory role through PPAR. See the table below for Studies and Clinical Trials.

Retinoid-Induced Irritation

This benefit is an exciting one. We know that Retinoids can cause dryness, barrier damage, irritation and even dermatitis. The exact mechanism isn't fully known, but some authors theorize that it has something to do with the dimerization of nuclear hormone receptors in the nucleus. For Retinoic acid to work, it has to bind RARs, which heterodimerize with RXRs. The number of RXRs in the nucleus is under tight control, and PPARs can bind RAR and RXR. (12) One interesting theory is that the side effects are due to RAR-RXR heterodimers that are not DNA-bound. PPAR may bind to RXR and reduce the non-DNA-bound RAR-RXR through direct competition.
In addition, PPAR's primary function is promoting barrier homeostasis and controlling inflammation. It could also just simply counteract the temporary barrier-damaging effects of retinol. In an interesting study, PPARa agonists WY-14643 and LXR agonist 22-Hydroxycholesterol were able to blunt the increase in TEWL caused by Retinoic acid. In addition, Retinoic acid treatment caused hyperproliferation and treatment with PPARa agonist reduced the proliferation. PPAR agonists also reduced retinoid-induced IL-1a, TNF-a and IL-8 mRNA expression (important mediators of retinoid dermatitis). (13) Another study showed that combining retinoic acid with a PPARa agonist increased the collagen content in the skin significantly: RA with 47%, 70% with 10-HSA, and 240% with a combination of both.

Studies and Clinical Trials

References:

1: Gupta, M., Mahajan, V., Mehta, K., Chauhan, P., & Rawat, R. (2015). Peroxisome proliferator-activated receptors (PPARs) and PPAR agonists: the “future” in dermatology therapeutics?. Archives Of Dermatological Research, 307(9), 767-780. https://doi.org/10.1007/s00403-015-1571-1

2. Rivier M, Safonova I, Lebrun P, Griffiths CE, Ailhaud G, Michel S. Differential expression of peroxisome proliferator-activated receptor subtypes during the differentiation of human keratinocytes. J Invest Dermatol 1998; 111:1116–1121.
3. Jiang, Y. J., Lu, B., Tarling, E. J., Kim, P., Man, M. Q., Crumrine, D., Edwards, P. A., Elias, P. M., & Feingold, K. R. (2010). Regulation of ABCG1 expression in human keratinocytes and murine epidermis. Journal of lipid research, 51(11), 3185–3195. https://doi.org/10.1194/jlr.M006445
4. Uchida, Y. (2014). Ceramide signaling in mammalian epidermis. Biochimica et Biophysica Acta, 1841(3), 453–462.
5. Rabionet, M., Gorgas, K., & Sandhoff, R. (2014). Ceramide synthesis in the epidermis. Biochimica et Biophysica Acta, 1841(3), 422–434.
6. Feingold, K. R. (2007). Thematic review series: skin lipids. The role of epidermal lipids in cutaneous permeability barrier homeostasis. Journal of Lipid Research, 48(12), 2531–2546.
7. Kim, Y., Park, K., Kim, J., Seo, H., Shin, K., & Lee, Y. et al. (2014). An Endoplasmic Reticulum Stress-Initiated Sphingolipid Metabolite, Ceramide-1-Phosphate, Regulates Epithelial Innate Immunity by Stimulating β-Defensin Production. Molecular And Cellular Biology, 34(24), 4368-4378. https://doi.org/10.1128/mcb.00599-14
8. Michalik, L., Desvergne, B., Tan, N. S., Basu-Modak, S., Escher, P., Rieusset, J., Peters, J. M., Kaya, G., Gonzalez, F. J., Zakany, J., Metzger, D., Chambon, P., Duboule, D., & Wahli, W. (2001). Impaired skin wound healing in peroxisome proliferator-activated receptor (PPAR)alpha and PPARbeta mutant mice. The Journal of cell biology, 154(4), 799–814. https://doi.org/10.1083/jcb.200011148
9. Delerive, P., Fruchart, J. C., & Staels, B. (2001). Peroxisome proliferator-activated receptors in inflammation control. The Journal of endocrinology, 169(3), 453–459. https://doi.org/10.1677/joe.0.1690453
10. Schmuth, M., Jiang, Y., Dubrac, S., Elias, P., & Feingold, K. (2008). Thematic Review Series: Skin Lipids. Peroxisome proliferator-activated receptors and liver X receptors in epidermal biology. Journal Of Lipid Research, 49(3), 499-509. https://doi.org/10.1194/jlr.r800001-jlr200)
11. Elias, P. M., & Feingold, K. R. (2005). Skin Barrier. CRC Press.
12. Source: Chan, L. S., & Wells, R. A. (2009). Cross-Talk between PPARs and the Partners of RXR: A Molecular Perspective. PPAR research, 2009, 925309. https://doi.org/10.1155/2009/925309
13. Kim, B., Kim, J. E., Kim, H., Lee, J. D., Choi, K. Y., & Lee, S. H. (2012). Co-treatment with retinyl retinoate and a PPARα agonist reduces retinoid dermatitis. International journal of dermatology, 51(6), 733–741. https://doi.org/10.1111/j.1365-4632.2011.05332.x
14. Kuenzli S, Saurat JH (2003) Peroxisome proliferator-activated receptors in cutaneous biology. Br J Dermatol 149:229–236
15. Kömüves, L. G., Hanley, K., Lefebvre, A. M., Man, M. Q., Ng, D. C., Bikle, D. D., Williams, M. L., Elias, P. M., Auwerx, J., & Feingold, K. R. (2000). Stimulation of PPARalpha promotes epidermal keratinocyte differentiation in vivo. The Journal of investigative dermatology, 115(3), 353–360. https://doi.org/10.1046/j.1523-1747.2000.00073.x
16. Kömüves, L. G., Hanley, K., Man, M. Q., Elias, P. M., Williams, M. L., & Feingold, K. R. (2000). Keratinocyte differentiation in hyperproliferative epidermis: topical application of PPARalpha activators restores tissue homeostasis. The Journal of investigative dermatology, 115(3), 361–367. https://doi.org/10.1046/j.1523-1747.2000.00076.x
17. Braissant O,WahliW (1998) Differential expression of peroxisome
proliferator-activated receptor-alpha-beta, and -gamma during rat
embryonic development. Endocrinology 139:2748–2754
18. Kuenzli S, Saurat JH (2003) Peroxisome proliferator-activated
receptors in cutaneous biology. Br J Dermatol 149:229–236
19. Westergaard M, Henningsen J, Svendsen ML, Johansen C, Jensen UB, Schroder HD et al (2001) Modulation of keratinocyte gene expression and differentiation by PPAR selective ligands and tetradecylthioacetic acid. J Invest Dermatol 116:702–712
20. Hanley K, Komuves LG, Bass NM, He SS, Jiang Y, Crumrine D et al (1999) Fetal epidermal differentiation and barrier development in vivo is accelerated by nuclear hormone receptor activators. J Invest Dermatol 113:788–795
21. Gupta, M., Mahajan, V., Mehta, K., Chauhan, P., & Rawat, R. (2015). Peroxisome proliferator-activated receptors (PPARs) and PPAR agonists: the ‘future’ in dermatology therapeutics?. Archives Of Dermatological Research, 307(9), 767-780. https://doi.org/10.1007/s00403-015-1571-1
22. Rivier, M., Safonova, I., Michel, S., Castiel, I., & Ailhaud, G. (2000). Peroxisome Proliferator-Activated Receptor-α Enhances Lipid Metabolism in a Skin Equivalent Model. Journal Of Investigative Dermatology, 114(4), 681-687. https://doi.org/10.1046/j.1523-1747.2000.00939.x
23. Chinetti G, Griglio S, Antonucci M, Torra IP, Delerive P, Majd Z, Fruchart JC,
Chapman J, Najib J, Staels B. Activation of proliferator-activated receptors alpha and gamma induces apoptosis of human monocyte-derived macrophages. J Biol Chem 1998; 273:25573–25580.
24. Kippenberger, S., Grundmann-Kollmann, M., Simon, S., Dang, T., Hardt-Weinelt, K., & Kaufmann, R. et al. (2001). Activators of Peroxisome Proliferator-Activated Receptors Protect Human Skin from Ultraviolet-B-Light-Induced Inflammation. Journal Of Investigative Dermatology, 117(6), 1430-1436. https://doi.org/10.1046/j.0022-202x.2001.01537.x
25. Rivier, M., Safonova, I., Michel, S., Castiel, I., & Ailhaud, G. (2000). Peroxisome Proliferator-Activated Receptor-α Enhances Lipid Metabolism in a Skin Equivalent Model. Journal Of Investigative Dermatology, 114(4), 681-687. https://doi.org/10.1046/j.1523-1747.2000.00939.x
26. Fluhr, J., Man, M., Hachem, J., Crumrine, D., Mauro, T., Elias, P., & Feingold, K. (2009). Topical Peroxisome Proliferator Activated Receptor Activators Accelerate Postnatal Stratum Corneum Acidification. Journal Of Investigative Dermatology, 129(2), 365-374. https://doi.org/10.1038/jid.2008.218
27. Rivier M, Safonova I, Lebrun P, Griffiths CE, Ailhaud G, Michel S. Differential expression of peroxisome proliferator-activated receptor subtypes during the differentiation of human keratinocytes. J Invest Dermatol 1998; 111:1116–1121.
28. Delerive, P., De Bosscher, K., Besnard, S., Vanden Berghe, W., Peters, J. M., Gonzalez, F. J., Fruchart, J. C., Tedgui, A., Haegeman, G., & Staels, B. (1999). Peroxisome proliferator-activated receptor alpha negatively regulates the vascular inflammatory gene response by negative cross-talk with transcription factors NF-kappaB and AP-1. The Journal of biological chemistry, 274(45), 32048–32054. https://doi.org/10.1074/jbc.274.45.32048
29. Sheu, M. Y., Fowler, A. J., Kao, J., Schmuth, M., Schoonjans, K., Auwerx, J., Fluhr, J. W., Man, M. Q., Elias, P. M., & Feingold, K. R. (2002). Topical peroxisome proliferator activated receptor-alpha activators reduce inflammation in irritant and allergic contact dermatitis models. The Journal of investigative dermatology, 118(1), 94–101. https://doi.org/10.1046/j.0022-202x.2001.01626.x
30. Su, C. G., Wen, X., Bailey, S. T., Jiang, W., Rangwala, S. M., Keilbaugh, S. A., Flanigan, A., Murthy, S., Lazar, M. A., & Wu, G. D. (1999). A novel therapy for colitis utilizing PPAR-gamma ligands to inhibit the epithelial inflammatory response. The Journal of clinical investigation, 104(4), 383–389. https://doi.org/10.1172/JCI7145
31. Lo Verme, J., Fu, J., Astarita, G., La Rana, G., Russo, R., Calignano, A., & Piomelli, D. (2005). The nuclear receptor peroxisome proliferator-activated receptor-alpha mediates the anti-inflammatory actions of palmitoylethanolamide. Molecular pharmacology, 67(1), 15–19. https://doi.org/10.1124/mol.104.006353
32. Delerive, P., Gervois, P., Fruchart, J. C., & Staels, B. (2000). Induction of IkappaBalpha expression as a mechanism contributing to the anti-inflammatory activities of peroxisome proliferator-activated receptor-alpha activators. The Journal of biological chemistry, 275(47), 36703–36707. https://doi.org/10.1074/jbc.M004045200
33. Elias, P. M., & Feingold, K. R. (2005). Skin Barrier. CRC Press.
34. Kim, B., Kim, J. E., Kim, H., Lee, J. D., Choi, K. Y., & Lee, S. H. (2012). Co-treatment with retinyl retinoate and a PPARα agonist reduces retinoid dermatitis. International journal of dermatology, 51(6), 733–741. https://doi.org/10.1111/j.1365-4632.2011.05332.x
35. Ramot, Y., Mastrofrancesco, A., Camera, E., Desreumaux, P., Paus, R., Picardo, M. (2015). The Role of PPARgamma-mediated signaling in skin biology and pathology: new target and opportunities for clinical dermatology. Experimental Dermatology, 24, 245-251. DOI: 10.1111/exd.12647
36. Man, M.-Q., Choi, E.-H., Schmuth, M., Crumrine, D., Uchida, Y., Elias, P. M., Holleran, W. M., & Feingold, K. R. (2006). Basis for Improved Permeability Barrier Homeostasis Induced by PPAR and LXR Activators: Liposensors Stimulate Lipid Synthesis, Lamellar Body Secretion, and Post-Secretory Lipid Processing. In Journal of Investigative Dermatology (Vol. 126, Issue 2, pp. 386–392). https://doi.org/10.1038/sj.jid.5700046
37. Chon, S. H., Tannahill, R., Yao, X., Southall, M. D., & Pappas, A. (2015). Keratinocyte differentiation and upregulation of ceramide synthesis induced by an oat lipid extract via the activation of PPAR pathways. Experimental dermatology, 24(4), 290–295. https://doi.org/10.1111/exd.12658
38. Schmuth M, Haqq CM, Cairns WJ, Holder JC, Dorsam S, Chang S et al (2004) Peroxisome proliferator-activated receptor (PPAR)-β/δ Stimulates differentiation and lipid accumulation in keratinocytes. J Invest Dermatol 122:971–983
39. Schmuth M, Moosbrugger-Martinz V, Blunder S, Dubrac S (2014) Role of PPAR, LXR, and PXR in epidermal homeostasis and inflammation. Biochim Biophys Acta 1841:463–473
40. Matsuura, H., Adachi, H., C. Smart, R., Xu, X., Arata, J., & M. Jetten, A. (1999). Correlation between expression of peroxisome proliferator-activated receptor β and squamous differentiation in epidermal and tracheobronchial epithelial cells. Molecular And Cellular Endocrinology, 147(1-2), 85-92. https://doi.org/10.1016/s0303-7207(98)00214-7
41. Rosenfield, R., Kentsis, A., Deplewski, D., & Ciletti, N. (1999). Rat Preputial Sebocyte Differentiation Involves Peroxisome Proliferator-Activated Receptors. Journal Of Investigative Dermatology, 112(2), 226-232. https://doi.org/10.1046/j.1523-1747.1999.00487.x
42. Schmuth M, Haqq CM, Cairns WJ, Holder JC, Dorsam S, Chang S, Lau P, Fowler AJ, Chuang G, Moser AH, Brown BE, Mao-Qiang M, Uchida Y, Schoonjans K, Auwerx J, Chambon P, Willson TM, Elias PM, Feingold KR. Peroxisome proliferator-activated receptor (PPAR)-beta/delta stimulates differentiation and lipid accumulation in keratinocytes. J Invest Dermatol 2004; 122:971–983.
43. Michalik L, Desvergne B, Tan NS, Basu-Modak S, Escher P, Rieusset J, Peters JM, Kaya G, Gonzalez FJ, Zakany J, Metzger D, Chambon P, Duboule D, Wahli W.
Impaired skin wound healing in peroxisome proliferator-activated receptor (PPAR) {alpha} and PPAR{beta} mutant mice. J Cell Biol 2001; 154:799–814.
44. Peters JM, Lee SS, Li W, Ward JM, Gavrilova O, Everett C, Reitman ML, Hudson LD, Gonzalez FJ. Growth, adipose, brain, and skin alterations resulting from targeted disruption
of the mouse peroxisome proliferator-activated receptor beta(delta). Mol Cell Biol 2000; 20:5119–5128.
45. Kuenzli S, Saurat JH. Effect of topical PPARbeta/delta and PPARgamma agonists on plaque psoriasis. A pilot study.. Dermatology 2003; 206:252–256.
46. Kuenzli S, Saurat JH. Peroxisome proliferator-activated receptors in cutaneous biology. Br J Dermatol 2003; 149:229–236.
47. Man MQ, Barish GD, Schmuth D, Crumrine Y, Barak S, Chang Y et al (2008) Deficiency of PPAR beta/delta in the epidermis results in defective cutaneous permeability barrier homeostasis and increased inflammation. J Invest Dermatol 128:370–377
48. Westergaard M, Henningsen J, Svendsen ML, Johansen C, Jensen UB, Schroder HD, Kratchmarova I, Berge RK, Iversen L, Bolund L, Kragballe K, Kristiansen K. Modulation of keratinocyte gene expression and differentiation by PPAR-selective ligands and tetradecylthioacetic acid. J Invest Dermatol 2001; 116:702–712
49. Tan NS, Michalik L, Noy N, Yasmin R, Pacot C, Heim M, Fluhmann B, Desvergne B, Wahli W. Critical roles of PPAR beta/delta in keratinocyte response to inflammation. Genes Dev 2001; 15:3263–3277.
50. Ricote, M., Li, A. C., Willson, T. M., Kelly, C. J., & Glass, C. K. (1998). The peroxisome proliferator-activated receptor-gamma is a negative regulator of macrophage activation. Nature, 391(6662), 79–82. https://doi.org/10.1038/34178
51. Jiang, Y. J., Lu, B., Kim, P., Paragh, G., Schmitz, G., Elias, P. M., & Feingold, K. R. (2008). PPAR and LXR activators regulate ABCA12 expression in human keratinocytes. The Journal of investigative dermatology, 128(1), 104–109. https://doi.org/10.1038/sj.jid.5700944
52. Westergaard, M., Henningsen, J., Svendsen, M. L., Johansen, C., Jensen, U. B., Schrøder, H. D., Kratchmarova, I., Berge, R. K., Iversen, L., Bolund, L., Kragballe, K., & Kristiansen, K. (2001). Modulation of keratinocyte gene expression and differentiation by PPAR-selective ligands and tetradecylthioacetic acid. The Journal of investigative dermatology, 116(5), 702–712. https://doi.org/10.1046/j.1523-1747.2001.01329.x
53. Wahli W. (2002). Peroxisome proliferator-activated receptors (PPARs): from metabolic control to epidermal wound healing. Swiss medical weekly, 132(7-8), 83–91.
54. Pascual G, Fong AL, Ogawa S, et al. A SUMOylation-dependent pathway mediates transrepression of inflammatory response genes by PPAR-gamma. Nature. 2005;437(7059):759-763. doi:10.1038/nature03988
55. Mao-Qiang, M., Fowler, A. J., Schmuth, M., Lau, P., Chang, S., Brown, B. E., Moser, A. H., Michalik, L., Desvergne, B., Wahli, W., Li, M., Metzger, D., Chambon, P. H., Elias, P. M., & Feingold, K. R. (2004). Peroxisome-proliferator-activated receptor (PPAR)-gamma activation stimulates keratinocyte differentiation. The Journal of investigative dermatology, 123(2), 305–312. https://doi.org/10.1111/j.0022-202X.2004.23235.x
56. Sugiyama H, Nonaka T, Kishimoto T et al. (2000) Peroxisome proliferator-activatedreceptors are expressed in human cultured mast cells: a possible role of these receptors in negative regulation of mast cell activation. Eur J Immunol 30: 3363-70
57. Marx N, Kehrle B, Kohlhammer K, et al. (2002) PPAR activators as antiinflammatory
mediators in human T lymphocytes: implications for atherosclerosis and transplantationassociated arteriosclerosis. Circ Res 90: 703-10
58. Henson P (2003) Suppression of macrophage inflammatory responses by PPARs. PNAS 100:6295-6
59. Man MQ, Choi EH, Schmuth M, et al. (2006) Basis for improved permeability barrier homeostasis induced by PPAR and LXR activators: liposensors stimulate lipid synthesis, lamellar body secretion, and post-secretory lipid processing. J Invest Dermatol 126: 386-92
60. Delerive P, Gervois P, Fruchart JC, et al. (2000). Induction of IκBα expression as a mechanism contributing to the anti-inflammatory activities of peroxisome proliferatoractivated receptor-α activators. J Biol Chem 275: 36703-7
61. Delerive P, Fruchart JC, Staels B (2001) Peroxisome proliferators-activated receptors in inflammation control. J Endocrinol 169: 453-9
62. Pascual G, Glass CK (2006) Nuclear receptor versus inflammation: mechanisms of transrepression. Trends Endocrinol Metab 17: 321-7