Inhibition of Cholesterol Biosynthesis Modulates Epithelial-Mesenchymal Transition in Primary Cicatricial Alopecia Through TGFβ and Angiotensin Receptors

Jump To References Section

Authors

  • Department of Zoology, University of Kerala, Thiruvananthapuram - 695581, Kerala ,IN
  • Department of Zoology, University of Kerala, Thiruvananthapuram - 695581, Kerala ,IN
  • Department of Zoology, University of Kerala, Thiruvananthapuram - 695581, Kerala ,IN
  • Department of Zoology, University of Kerala, Thiruvananthapuram - 695581, Kerala ,IN
  • Department of Zoology, University of Kerala, Thiruvananthapuram - 695581, Kerala ,IN

DOI:

https://doi.org/10.18311/jer/2023/33509

Keywords:

Cholesterol, Fibrosis, Hair, PPARγ, Pioglitazone, Transfection

Abstract

Introduction: Primary Cicatricial Alopecia (PCA) is an autoimmune condition that affects the skin and causes hair loss in patients. In PCA the hair follicles of the patients are irreversibly damaged and replaced with fibrous tissue. This diseased condition lends relevance to our work since the fibrosis raises the potential that PCA may be affected in some way by the Epithelial Mesenchymal Transition (EMT). We used small interfering RNAs (siRNA) of TGFβ, AGTR and their regulators to identify the EMT modulation. Because these molecules mediate the induction of EMT. This study explores the idea of lowering PCA fibrosis by modifying EMT markers. Methods: We chose 7 DHC and BM15766 to investigate the function of cholesterol biosynthesis inhibition. We employed the HFORS in vitro and the mouse in vivo model system to examine EMT regulation PCA. Quantitative real-time PCR was utilised to examine the expression of genes in PCA scalp samples, compound-treated HFORS, and mouse tissues; immunohistochemistry was used to confirm the protein estimate in the scalp samples; and small interfering RNA (siRNA) transfection was used to identify the functional analysis of TGFβ and AGTR. Results: Reduced cholesterol production in PCA patients leads to permanent hair follicle damage. The in vitro and in vivo study using 7DHC and BM15766 revealed cells were positive for the EMT markers. PPARγ, AhR, and AGTR together can act as vital EMT regulators. As a result, the PPARγ agonist, AhR, and AGTR antagonist significantly downregulate the expression of CDH1, SNAIL1, and SMA. The markers of EMT are likewise deregulated by the transfection of siRNA for TGFβ and AGTR. Conclusion: We clarify how EMT is regulated in hair loss circumstances by suppressing cholesterol biosynthesis. We further confirm that EMT modulators (PPARγ, AhR, AGTR, and TGFβ) and siRNA can be employed as potentially effective strategies to slow the advancement of EMT. As a result, we propose these cholesterol and EMT modulators as potential inhibitors in PCA etiology.

Downloads

Download data is not yet available.

Metrics

Metrics Loading ...

Downloads

Published

2023-05-12

How to Cite

Nikhila, L., Surya, S., Najeeb, S. H., Binumon, T. M., & Sreejith, P. P. (2023). Inhibition of Cholesterol Biosynthesis Modulates Epithelial-Mesenchymal Transition in Primary Cicatricial Alopecia Through TGFβ and Angiotensin Receptors. Journal of Endocrinology and Reproduction, 27(1), 40–53. https://doi.org/10.18311/jer/2023/33509

Issue

Section

Original Research

 

References

Harries MJ, Paus R. The pathogenesis of primary cicatricial alopecias. The American Journal of Pathology. 2010; 177(5):2152-62. https://doi.org/10.2353/ ajpath.2010.100454 PMid: 20889564 PMCid: PMC2966773 DOI: https://doi.org/10.2353/ajpath.2010.100454

Panicker SP, Ganguly T, Consolo M,, et al. Sterol intermediates of cholesterol biosynthesis inhibit hair growth and trigger an innate immune response in cicatricial alopecia. PLoS One. 2012; 7(6):e38449. https:// doi.org/10.1371/journal.pone.0038449 PMid:22685570 PMCid: PMC3369908 DOI: https://doi.org/10.1371/journal.pone.0038449

Karnik P, Tekeste Z, McCormick TS,, et al. Hair follicle stem cell-specific PPARγ deletion causes scarring alopecia. Journal of Investigative Dermatology. 2009; 129(5):1243-57. https://doi.org/10.1038/jid.2008.369 PMid:19052558 PMCid:PMC3130601

Harries MJ, Trueb RM, Tosti A,, et al. How not to get scar (r) ed: pointers to the correct diagnosis in patients with suspected primary cicatricial alopecia. British Journal of Dermatology. 2009; 160(3):482-501. https://doi. org/10.1111/j.1365-2133.2008.09008.x PMid:19183169 DOI: https://doi.org/10.1111/j.1365-2133.2008.09008.x

Chen X, Liu B, Li Y,, et al. Dihydrotestosterone regulates hair growth through the Wnt/β-catenin pathway in C57BL/6 mice and in vitro organ culture. Frontiers in Pharmacology. 2020; 10:1528. https://doi. org/10.3389/fphar.2019.01528 PMid:32038233 PMCid: PMC6989660 DOI: https://doi.org/10.3389/fphar.2019.01528

Tavakolpour S, Mahmoudi H, Abedini R,, et al. Frontal fibrosing alopecia: An update on the hypothesis of pathogenesis and treatment. International Journal of Women’s Dermatology. 2019; 5(2):116-23. https:// doi.org/10.1016/j.ijwd.2018.11.003 PMid:30997385 PMCid:PMC6451751 DOI: https://doi.org/10.1016/j.ijwd.2018.11.003

Mirmirani P, Karnik P. Lichen planopilaris treated with a peroxisome proliferator-activated receptor γ agonist. Archives of Dermatology. 2009; 145(12):1363-6. https://doi.org/10.1001/archdermatol.2009.283 PMid: 20026843 PMCid:PMC2937154

Schaffrin-Nabe D, Schmitz I, Josten-Nabe A,, et al. The influence of various parameters on the success of sensor-controlled scalp cooling in preventing chemotherapy-induced alopecia. Oncology Research and Treatment. 2015; 38(10):489-95. https://doi. org/10.1159/000440636 PMid:26451590 DOI: https://doi.org/10.1159/000440636

Harries MJ, Sinclair RD, Macdonald‐Hull S,, et al. Management of primary cicatricial alopecias: options for treatment. British Journal of Dermatology. 2008; 159(1):1-22. https://doi.org/10.1111/j.1365- 2133.2008.08591.x PMid: 18489608 DOI: https://doi.org/10.1111/j.1365-2133.2008.08591.x

Mirmirani P, Willey A, Headington JT,, et al. Primary cicatricial alopecia: histopathologic findings do not distinguish clinical variants. Journal of the American Academy of Dermatology. 2005; 52(4):637-43. https:// doi.org/10.1016/j.jaad.2004.07.069 PMid:15793514 DOI: https://doi.org/10.1016/j.jaad.2004.07.069

MacDonald A, Clark C, Holmes S. Frontal fibrosing alopecia: a review of 60 cases. Journal of the American Academy of Dermatology. 2012; 67(5):955-61. https:// doi.org/10.1016/j.jaad.2011.12.038 PMid:22503342 DOI: https://doi.org/10.1016/j.jaad.2011.12.038

Najeeb SH, Binumon TM, Surya S, Nikhila L, Sreejith PP. Cholesterolgenic Inhibition causes permanent hair follicle damage by activating fibrosis via the angiotensin receptor. Journal of Endocrinology and Reproduction. 2022; 26(3):187-204. https://doi. org/10.25303/2604rjce1116

Imanishi H, Ansell DM, Chéret J,, et al. Epithelialto- mesenchymal stem cell transition in a human organ: lessons from lichen planopilaris. Journal of Investigative Dermatology. 2018; 138(3):511-9. https:// doi.org/10.1016/j.jid.2017.09.047 PMid:29106928 DOI: https://doi.org/10.1016/j.jid.2017.09.047

Hibino T, Nishiyama T. Role of TGF-β2 in the human hair cycle. Journal of Dermatological Science. 2004; 35(1):9-18. https://doi.org/10.1016/j.jdermsci. 2003.12.003 PMid: 15194142 DOI: https://doi.org/10.1016/j.jdermsci.2003.12.003

Karnik P, Tekeste Z, McCormick TS,, et al. Hair follicle stem cell-specific PPARγ deletion causes scarring alopecia. Journal of Investigative Dermatology. 2009; 129(5):1243-57. https://doi.org/10.1038/jid.2008.369 PMid:19052558 PMCid:PMC3130601 DOI: https://doi.org/10.1038/jid.2008.369

Aoki Y, Maeno T, Aoyagi K,, et al. Pioglitazone, a peroxisome proliferator-activated receptor gamma ligand, suppresses bleomycin-induced acute lung injury and fibrosis. Respiration. 2009; 77(3):311-9. https://doi. org/10.1159/000168676 PMid:18974632 DOI: https://doi.org/10.1159/000168676

Ramot Y, Mastrofrancesco A, Camera E,, et al. The role of PPAR γ‐mediated signalling in skin biology and pathology: New targets and opportunities for clinical dermatology. Experimental Dermatology. 2015; 24(4):245-51. https://doi.org/10.1111/exd.12647 PMid:25644500

Henson P. Suppression of macrophage inflammatory responses by PPARs. Proceedings of the National Academy of Sciences. 2003; 100(11):6295-6. https://doi. org/10.1073/pnas.1232410100 PMid:12756292 PMCid: PMC164439 DOI: https://doi.org/10.1073/pnas.1232410100

Marx N, Kehrle B, Kohlhammer K,, et al. PPAR activators as antiinflammatory mediators in human T lymphocytes: implications for atherosclerosis and transplantation-associated arteriosclerosis. Circulation Research. 2002; 90(6):703-10. https://doi. org/10.1161/01.RES.0000014225.20727.8F PMid: 11934839 PMCid:PMC4231718 DOI: https://doi.org/10.1161/01.RES.0000014225.20727.8F

Sugiyama H, Nonaka T, Kishimoto T,, et al. Peroxisome proliferator‐activated receptors are expressed in human cultured mast cells: A possible role of these receptors in negative regulation of mast cell activation. European Journal of Immunology. 2000; 30(12):3363-70. https://doi. org/10.1002/1521-4141(2000012)30:12<3363::AIDIMMU3363> 3.0.CO;2-B PMid:11093153 DOI: https://doi.org/10.1002/1521-4141(2000012)30:12<3363::AID-IMMU3363>3.0.CO;2-B

Gamret AC, Potluri VS, Krishnamurthy K, Fertig RM. Frontal fibrosing alopecia: Efficacy of treatment modalities. International Journal of Women’s Health. 2019; 11:273. https://doi.org/10.2147/IJWH.S177308 PMid:31118828 PMCid:PMC6500869 DOI: https://doi.org/10.2147/IJWH.S177308

Noakes R. Frontal fibrosing alopecia. An example of disrupted aryl hydrocarbon receptor-mediated immunological homeostasis in the skin? Clinical, Cosmetic and Investigational Dermatology. 2020; 13:479. https:// doi.org/10.2147/CCID.S262803 PMid:32801823 PMCid: PMC7399449 DOI: https://doi.org/10.2147/CCID.S262803

Bonefeld-Jørgensen EC, Long M, Hofmeister MV, Vinggaard AM. Endocrine-disrupting potential of bisphenol A, bisphenol A dimethacrylate, 4-n-nonylphenol, and 4-n-octylphenol in vitro: New data and a brief review. Environmental Health Perspectives. 2007; 115(Suppl 1):69-76. https://doi.org/10.1289/ehp.9368 PMid:18174953 PMCid:PMC2174402

Nakamura M, Tokura Y. Epithelial-mesenchymal transition in the skin. Journal of Dermatological Science. 2011; 61(1):7-13. https://doi.org/10.1016/j.jdermsci. 2010.11.015 PMid:21167690 DOI: https://doi.org/10.1016/j.jdermsci.2010.11.015

Nakamura M, Tokura Y. Expression of Snail1 in the fibrotic dermis of postmenopausal frontal fibrosing alopecia: Possible involvement of an epithelial-mesenchymal transition and a review of the Japanese patients. British Journal of Dermatology. 2010; 162(5):1152- 4. https://doi.org/10.1111/j.1365-2133.2010.09682.x PMid:20132204 DOI: https://doi.org/10.1111/j.1365-2133.2010.09682.x

Lamouille S, Xu J, Derynck R. Molecular mechanisms of epithelial-mesenchymal transition. Nature Reviews Molecular Cell Biology. 2014; 15:178-96. https://doi.org/10.1038/nrm3758 PMid:24556840 PMCid: PMC4240281 DOI: https://doi.org/10.1038/nrm3758

Skrypek N, Goossens S, De Smedt E,, et al. Epithelial-to-mesenchymal transition: Epigenetic reprogramming driving cellular plasticity. Trends in Genetics. 2017; 33(12):943-59. https://doi. org/10.1016/j.tig.2017.08.004 PMid:28919019 DOI: https://doi.org/10.1016/j.tig.2017.08.004

Singh M, Yelle N, Venugopal C, Singh SK. EMT: Mechanisms and therapeutic implications. Pharmacology and Therapeutics. 2018; 182:80-94. https://doi.org/10.1016/j.pharmthera.2017.08.009 PMid: 28834698 DOI: https://doi.org/10.1016/j.pharmthera.2017.08.009

Scanlon CS, Van Tubergen EA, Inglehart RC, D’silva NJ. Biomarkers of epithelial-mesenchymal transition in squamous cell carcinoma. Journal of Dental Research. 2013; 92(2):114-21. https://doi. org/10.1177/0022034512467352 PMid:23128109 PMCid:PMC3545688 DOI: https://doi.org/10.1177/0022034512467352

Jamora C, Lee P, Kocieniewski P,, et al. A signaling pathway involving TGF-β2 and snail in hair follicle morphogenesis. PLoSBiology. 2005; 3(1):e11. https:// doi.org/10.1371/journal.pbio.0030011 PMid:15630473 PMCid:PMC539061 DOI: https://doi.org/10.1371/journal.pbio.0030011

Franci C, Takkunen M, Dave N,, et al. Expression of Snail protein in tumor-stroma interface. Oncogene. 2006; 25(37):5134-44. https://doi.org/10.1038/ sj.onc.1209519 PMid:16568079 DOI: https://doi.org/10.1038/sj.onc.1209519

Levy V, Lindon C, Zheng Y,, et al. Epidermal stem cells arise from the hair follicle after wounding. The FASEB Journal. 2007; 21(7):1358-66. https://doi.org/10.1096/ fj.06-6926com PMid:17255473 DOI: https://doi.org/10.1096/fj.06-6926com

Yang H, Adam RC, Ge Y,, et al. Epithelial-mesenchymal micro-niches govern stem cell lineage choices. Cell. 2017; 169(3):483-96. https://doi.org/10.1016/j. cell.2017.03.038 PMid:28413068 PMCid:PMC5510744 DOI: https://doi.org/10.1016/j.cell.2017.03.038

Veraitch O, Kobayashi T, Imaizumi Y,, et al. Human induced pluripotent stem cell-derived ectodermal precursor cells contribute to hair follicle morphogenesis In Vivo. Journal of Investigative Dermatology. 2013; 133(6):1479-88. https://doi.org/10.1038/jid.2013.7 PMid:23321923 DOI: https://doi.org/10.1038/jid.2013.7

Neilson EG. Mechanisms of disease: Fibroblasts- A new look at an old problem. Nature Clinical Practice Nephrology. 2006; 2(2):101-8. https://doi.org/10.1038/ ncpneph0093 PMid:16932401 DOI: https://doi.org/10.1038/ncpneph0093

Nikitorowicz-Buniak J, Denton CP, Abraham D, Stratton R. Partially evoked Epithelial-Mesenchymal Transition (EMT) is associated with increased TGFβ signaling within lesional scleroderma skin. PloS one. 2015; 10(7):e0134092. https://doi.org/10.1371/journal. pone.0134092 PMid:26217927 PMCid:PMC4517793 DOI: https://doi.org/10.1371/journal.pone.0134092

Sonnylal S, Xu S, Jones H,, et al. Connective tissue growth factor causes EMT-like cell fate changes in vivo and in vitro. Journal of Cell Science. 2013; 126(10):2164-75. https://doi.org/10.1242/jcs.111302 PMid:23525012 PMCid:PMC3672936 DOI: https://doi.org/10.1242/jcs.111302

Yao Y, Li Y, Zeng X,, et al. Losartan alleviates renal fibrosis and inhibits Endothelial-To-Mesenchymal Transition (EMT) under high-fat diet-induced hyperglycemia. Frontiers in Pharmacology. 2018; 9:1213. https:// doi.org/10.3389/fphar.2018.01213 PMid:30420805 PMCid:PMC6215973 DOI: https://doi.org/10.3389/fphar.2018.01213

Wylie-Sears J, Levine RA, Bischoff J. Losartan inhibits endothelial-to-mesenchymal transformation in mitral valve endothelial cells by blocking transforming growth factor-β-induced phosphorylation of ERK. Biochemical and Biophysical Research Communications. 2014; 446(4):870-5. https://doi.org/10.1016/j.bbrc.2014.03.014 PMid:24632204 PMCid: PMC4007266 DOI: https://doi.org/10.1016/j.bbrc.2014.03.014

deCarvalho LT, Izhakoff N, Meah N, Sinclair R. Alopecia areata mimicking frontal fibrosing alopecia. Cureus. 2021; 13(2):e13361.

Bonefeld-Jørgensen EC, Long M, Hofmeister MV, Vinggaard AM. Endocrine-disrupting potential of bisphenol A, bisphenol A dimethacrylate, 4-n-nonylphenol, and 4-n-octylphenol in vitro: New data and a brief review. Environmental Health Perspectives. 2007; 115(Suppl 1):69-76. https://doi.org/10.1289/ehp.9368 PMid:18174953 PMCid:PMC2174402 DOI: https://doi.org/10.1289/ehp.9368

Yan L, Cao R, Wang L,, et al. Epithelial-mesenchymal transition in keloid tissues and TGF‐β1-induced hair follicle outer root sheath keratinocytes. Wound Repair and Regeneration. 2015; 23(4):601-10. https://doi. org/10.1111/wrr.12320 PMid:26036684 DOI: https://doi.org/10.1111/wrr.12320

Mirmirani P, Karnik P. Lichen planopilaris treated with a peroxisome proliferator-activated receptor γ agonist. Archives of Dermatology. 2009; 145(12):1363- 6. https://doi.org/10.1001/archdermatol.2009.283 PMid:20026843 PMCid:PMC2937154 DOI: https://doi.org/10.1001/archdermatol.2009.283

Harries MJ, Jimenez F, Izeta A,, et al. Lichen planopilaris and frontal fibrosing alopecia as model epithelial stem cell diseases. Trends in molecular Medicine. 2018; 24(5):435-48. https://doi.org/10.1016/j. molmed.2018.03.007 PMid: 29661566 DOI: https://doi.org/10.1016/j.molmed.2018.03.007

Price VH. The medical treatment of cicatricial alopecia. In: Seminars in Cutaneous Medicine and Surgery. 2006; 25(1):56-59. https://doi.org/10.1016/j.sder.2006.01.008 PMid:16616304 DOI: https://doi.org/10.1016/j.sder.2006.01.008

Mesinkovska NA, Tellez A, Dawes D,, et al. The use of oral pioglitazone in the treatment of lichen planopilaris. Journal of the American Academy of Dermatology. 2015; 72(2):355-6. https://doi.org/10.1016/j.jaad.2014.10.036 PMid: 25592345 DOI: https://doi.org/10.1016/j.jaad.2014.10.036

Ramot Y, Mastrofrancesco A, Camera E,, et al. The role of PPAR γ‐mediated signalling in skin biology and pathology: new targets and opportunities for clinical dermatology. Experimental Dermatology. 2015; 24(4):245-51. https://doi.org/10.1111/exd.12647 PMid:25644500 DOI: https://doi.org/10.1111/exd.12647

Rannug A, Rannug U, Rosenkranz HS,, et al. Certain photooxidized derivatives of tryptophan bind with very high affinity to the Ah receptor and are likely to be endogenous signal substances. Journal of Biological Chemistry. 1987; 262(32):15422-7. https://doi. org/10.1016/S0021-9258(18)47743-5 PMid:2824460 DOI: https://doi.org/10.1016/S0021-9258(18)47743-5

Rico-Leo EM, Alvarez-Barrientos A, Fernandez- Salguero PM. Dioxin receptor expression inhibits basal and transforming growth factor β-induced epithelial- to-mesenchymal transition. Journal of Biological Chemistry. 2013; 288(11):7841-56. https:// doi.org/10.1074/jbc.M112.425009 PMid:23382382 PMCid:PMC3597822 DOI: https://doi.org/10.1074/jbc.M112.425009

Van Den Bogaard EH, Podolsky MA, Smits JP,, et al. Genetic and pharmacological analysis identifies a physiological role for the AHR in epidermal differentiation. Journal of Investigative Dermatology. 2015; 135(5):1320-8. https://doi.org/10.1038/jid.2015.6 PMid: 25602157 PMCid:PMC4402116 DOI: https://doi.org/10.1038/jid.2015.6

Cai Y, Shen X, Ding C,, et al. Pivotal role of dermal IL-17-producing γδ T cells in skin inflammation. Immunity. 2011; 35(4):596-610. https://doi. org/10.1016/j.immuni.2011.10.006 PMid:21982596 PMCid:PMC3205267 DOI: https://doi.org/10.1016/j.immuni.2011.08.001

Qiu J, Heller JJ, Guo X,, et al. The aryl hydrocarbon receptor regulates gut immunity through modulation of innate lymphoid cells. Immunity. 2012; 36(1):92- 104. https://doi.org/10.1016/j.immuni.2011.11.011 PMid:22177117 PMCid:PMC3268875 DOI: https://doi.org/10.1016/j.immuni.2011.11.011

AlQudah M, Hale TM., Czubryt MP. Targeting the renin-angiotensin-aldosterone system in fibrosis. Matrix Biology. 2020; 91:92-108. https://doi.org/10.1016/j.matbio.2020.04.005 PMid:32422329 PMCid:PMC7434656 DOI: https://doi.org/10.1016/j.matbio.2020.04.005

Pallasch FB, Schumacher U. Angiotensin inhibition, TGF-β and EMT in cancer. Cancers. 2020; 12(10):2785. https://doi.org/10.3390/cancers12102785 PMid:32998363 PMCid:PMC7601465 DOI: https://doi.org/10.3390/cancers12102785

Chen J, Chen JK, Harris RC. Angiotensin II induces epithelial-to-mesenchymal transition in renal epithelial cells through reactive oxygen species/Src/caveolinmediated activation of an epidermal growth factor receptor-extracellular signal-regulated kinase signaling pathway. Molecular and Cellular Biology. 2012; 32(5):981-91. https://doi.org/10.1128/MCB.06410-11 PMid:22215616 PMCid:PMC3295195 DOI: https://doi.org/10.1128/MCB.06410-11

Most read articles by the same author(s)