Please wait a minute...
Frontiers of Medicine

ISSN 2095-0217

ISSN 2095-0225(Online)

CN 11-5983/R

Postal Subscription Code 80-967

2018 Impact Factor: 1.847

Front. Med.    2020, Vol. 14 Issue (3) : 305-317    https://doi.org/10.1007/s11684-019-0722-8
RESEARCH ARTICLE
Keratin 5-Cre-driven deletion of Ncstn in an acne inversa-like mouse model leads to a markedly increased IL-36a and Sprr2 expression
Jun Yang1, Lianqing Wang1,2, Yingzhi Huang1, Keqiang Liu1, Chaoxia Lu1, Nuo Si1, Rongrong Wang1, Yaping Liu1(), Xue Zhang1()
1. McKusick-Zhang Center for Genetic Medicine, State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China
2. Center of Translational Medicine, Central Hospital of Zibo, Shandong University, Zibo 255036, China
 Download: PDF(7688 KB)   HTML
 Export: BibTeX | EndNote | Reference Manager | ProCite | RefWorks
Abstract

Familial acne inversa (AI) is an autoinflammatory disorder that affects hair follicles and is caused by loss-of-function mutations in g-secretase component genes. We and other researchers showed that nicastrin (NCSTN) is the most frequently mutated gene in familial AI. In this study, we generated a keratin 5-Cre-driven epidermis-specific Ncstn conditional knockout mutant in mice. We determined that this mutant recapitulated the major phenotypes of AI, including hyperkeratosis of hair follicles and inflammation. In Ncstnflox/flox;K5-Cre mice, the IL-36a expression level markedly increased starting from postnatal day 0 (P0), and this increase occurred much earlier than those of TNF-α, IL-23A, IL-1b, and TLR4. RNA-Seq analysis indicated that Sprr2d, a member of the small proline-rich protein 2 family, in the skin tissues of the Ncstnflox/flox;K5-Cre mice was also upregulated on P0. Quantitative reverse-transcription polymerase chain reaction showed that other Sprr2 genes had a similar expression pattern. Our findings suggested that IL-36a might be a key inflammatory cytokine in the pathophysiology of AI and implicate malfunction of the skin barrier in the pathogenesis of AI.

Keywords acne inversa mouse model      interleukin 1 family, member 6      small proline rich protein 2D      key inflammatory cytokine     
Corresponding Author(s): Yaping Liu,Xue Zhang   
Just Accepted Date: 21 November 2019   Online First Date: 25 December 2019    Issue Date: 08 June 2020
 Cite this article:   
Jun Yang,Lianqing Wang,Yingzhi Huang, et al. Keratin 5-Cre-driven deletion of Ncstn in an acne inversa-like mouse model leads to a markedly increased IL-36a and Sprr2 expression[J]. Front. Med., 2020, 14(3): 305-317.
 URL:  
https://academic.hep.com.cn/fmd/EN/10.1007/s11684-019-0722-8
https://academic.hep.com.cn/fmd/EN/Y2020/V14/I3/305
Fig.1  Mouse mutant construction and verification. (A) Conditional Ncstn knockout strategy. (B) Primers used for genotyping the mice. (C) Agarose gel results showing the genotypes of the mice. K5-Cre mice were named WT. The cKO/cKO group was Ncstnflox/flox;K5-Cre. The cKO/+ group corresponded to Ncstnflox/+;K5-Cre. M, DNA marker (DL2000). (D) The knockout efficiency of Ncstn in the skin of WT and Ncstnflox/flox;K5-Cre mice was determined at the mRNA level on P0 and P30. Data shown were mean±standard deviation (SD) of three independent experiments, and each experiment was performed in duplicate. Two asterisks denoted P<0.01. The ratio of Ncstn to b-actin in WT mice was set to 1. The (E) presence/absence and (F) quantification level of Ncstn protein in WT and Ncstnflox/flox;K5-Cre mice skin were determined on P30. (G) Immunofluorescence staining results for Ncstn (green) and keratin 5 (red) in the skin of WT (a–c) and Ncstnflox/flox;K5-Cre (d–f) mice on P30. Scale bar= 50? mm.
Fig.2  Phenotypic and pathophysiologic characterization of the mouse model. (A) The skin appearance of Ncstnflox/flox;K5-Cre and WT mice at the indicated postnatal ages. (B) Hematoxylin- and eosin-stained skin sections on P15, P30, and P45. Scale bar= 50 mm. (C) mRNA expression of Notch1 in the skin of WT and Ncstnflox/flox;K5-Cre mice on P0 and P30, ascertained by qRT-PCR. Data shown were mean±SD of three experiments, and each experiment was performed in duplicate. The ratio of Notch1 to b-actin in WT mice on P0 was set to 1. Two asterisks denoted P<0.01. Western blot was used to determine the (D) presence/absence and (E) quantification level of the cleaved Notch1 (NICD) in the mouse skin. Results of quantitative reverse-transcription polymerase chain reaction for (F) Il23a, (G) Tnf-a, (H) Il1b, and (I) Tlr4. Data shown were mean±SD of three experiments, and each experiment was performed in duplicate. The ratio of the gene of interest to b-actin in WT mice on P0 was set to 1.
Fig.3  Result of microarray analysis of Ncstnflox/flox;K5-Cre mice. (A) The scatter plot on P0, P10, P15, P30, and P45 and (B) the hierarchical clustering map. (C) Three genes were screened from the intersection of four sets of data (P0, P10, P15, and P30) and intersected with the fifth set of data (P45), leaving only two differentially expressed genes.
Fig.4  Upregulation of IL-36a and LCN2 in the skin of the mouse model. Results of the quantitative reverse-transcription polymerase chain reaction and Western blot analysis of (A, B) IL-36a and (C, D) LCN2 expression in the skin of WT and Ncstnflox/flox;K5-Cre mice on P0, P15, P30, and P45. Data shown were mean±SD of three independent experiments, and each experiment was performed in duplicate. The ratio of the gene of interest to b-actin in WT mice on P0 was set to 1. Immunohistochemistry results for (E) CD68 (a marker of macrophages) and CD11c (a marker of dendritic cells), (F) IL-36a and keratin 5 (markers of keratinocytes), (G) RORgt (a marker of Th17 cells and ILC3) and IL-17A, and (H) TNF-a on P30 in the skin of WT and Ncstnflox/flox;K5-Cre mice. Arrows specify the positively stained areas. Scale bar= 50 mm. The cKO/cKO group corresponded to Ncstnflox/flox;K5-Cre mice.
Fig.5  RNA-seq analysis of Ncstnflox/flox;K5-Cre mice versus K5-cre mice on P0 and P30. (A) Heat clustering map; (B,C) enriched pathways; (D,E) enriched gene ontology terms; and (F,G) volcano plot.
Fig.6  Increased Sprr2dexpression in the skin of the AI mouse model. The mRNA expression level of the Sprr2 gene cluster in the skin of Ncstnflox/flox;K5-Cre mice compared with that of WT mice at four stages (A–D). The ratio of the gene of interest to b-actin in WT mice on P0 was set to 1. Data shown were mean±SD of three experiments, and each experiment was performed in duplicate. The cKO/cKO group corresponded to Ncstnflox/flox;K5-Cre mice.
Fig.7  Upregulation of SPRR2 in both NCSTN- and NOTCH1-downregulated HaCaT cells. siRNA-induced downregulation of (A, D) mRNA and (B, E) protein expression levels of NCSTN and NOTCH1 was depicted in HaCaT cells. Two asterisks denoted P<0.01. The ratio of the gene of interest to b-actin in the blank control was set to 1. The mRNA expression levels of IL-36a, LCN2, and SPRR2 were measured through quantitative reverse-transcription polymerase chain reaction in HaCaT cells in which (C) NCSTN or (F) NOTCH1 was knocked down. The ratio of the gene of interest to b-actin in the negative control was set to 1. Data shown were mean±SD of three experiments, and each experiment was performed in duplicate. NC, negative control. Blank, blank control.
1 J Revuz. Hidradenitis suppurativa. J Eur Acad Dermatol Venereol 2009; 23(9): 985–998
https://doi.org/10.1111/j.1468-3083.2009.03356.x pmid: 19682181
2 A Garg, JS Kirby, J Lavian, G Lin, A Strunk. Sex- and age-adjusted population analysis of prevalence estimates for hidradenitis suppurativa in the United States. JAMA Dermatol 2017; 153(8): 760–764
https://doi.org/10.1001/jamadermatol.2017.0201 pmid: 28492923
3 JE Revuz, F Canoui-Poitrine, P Wolkenstein, C Viallette, G Gabison, F Pouget, F Poli, O Faye, JC Roujeau, G Bonnelye, JJ Grob, S Bastuji-Garin. Prevalence and factors associated with hidradenitis suppurativa: results from two case-control studies. J Am Acad Dermatol 2008; 59(4): 596–601
https://doi.org/10.1016/j.jaad.2008.06.020 pmid: 18674845
4 C Coughlan, W Ledger, Q Wang, F Liu, A Demirol, T Gurgan, R Cutting, K Ong, H Sallam, TC Li. Recurrent implantation failure: definition and management. Reprod Biomed Online 2014; 28(1): 14–38
https://doi.org/10.1016/j.rbmo.2013.08.011 pmid: 24269084
5 GB Jemec, M Heidenheim, NH Nielsen. The prevalence of hidradenitis suppurativa and its potential precursor lesions. J Am Acad Dermatol 1996; 35(2 Pt 1): 191–194
https://doi.org/10.1016/S0190-9622(96)90321-7 pmid: 8708018
6 JR Ingram. The genetics of hidradenitis suppurativa. Dermatol Clin 2016; 34(1): 23–28
https://doi.org/10.1016/j.det.2015.07.002 pmid: 26617354
7 JV Schmitt, G Bombonatto, M Martin, HA Miot. Risk factors for hidradenitis suppurativa: a pilot study. An Bras Dermatol 2012; 87(6): 936–938
https://doi.org/10.1590/S0365-05962012000600024 pmid: 23197222
8 I Brook, EH Frazier. Aerobic and anaerobic microbiology of axillary hidradenitis suppurativa. J Med Microbiol 1999; 48(1): 103–105
https://doi.org/10.1099/00222615-48-1-103 pmid: 9920133
9 I Jansen, P Altmeyer, G Piewig. Acne inversa (alias hidradenitis suppurativa). J Eur Acad Dermatol Venereol 2001; 15(6): 532–540
https://doi.org/10.1046/j.1468-3083.2001.00303.x pmid: 11843212
10 M Gao, PG Wang, Y Cui, S Yang, YH Zhang, D Lin, KY Zhang, YH Liang, LD Sun, KL Yan, FL Xiao, W Huang, XJ Zhang. Inversa acne (hidradenitis suppurativa): a case report and identification of the locus at chromosome 1p21.1-1q25.3. J Invest Dermatol 2006; 126(6): 1302–1306
https://doi.org/10.1038/sj.jid.5700272 pmid: 16543891
11 JM Von Der Werth, HC Williams, JA Raeburn. The clinical genetics of hidradenitis suppurativa revisited. Br J Dermatol 2015; 142: 947–953
https://doi.org/DOI: 10.1046/j.1365-2133.2000.03476.x
12 Y Nomura, T Nomura, S Suzuki, M Takeda, O Mizuno, Y Ohguchi, R Abe, Y Murata, H Shimizu. A novel NCSTN mutation alone may be insufficient for the development of familial hidradenitis suppurativa. J Dermatol Sci 2014; 74(2): 180–182
https://doi.org/10.1016/j.jdermsci.2014.01.013 pmid: 24581508
13 B Wang, W Yang, W Wen, J Sun, B Su, B Liu, D Ma, D Lv, Y Wen, T Qu, M Chen, M Sun, Y Shen, X Zhang. γ-Secretase gene mutations in familial acne inversa. Science 2010; 330(6007): 1065
https://doi.org/10.1126/science.1196284 pmid: 20929727
14 RJ Kelleher 3rd, J Shen. Genetics. γ-Secretase and human disease. Science 2010; 330(6007): 1055–1056
https://doi.org/10.1126/science.1198668 pmid: 21097925
15 E Prens, I Deckers. Pathophysiology of hidradenitis suppurativa: an update. J Am Acad Dermatol 2015; 73(5 Suppl 1): S8–S11
https://doi.org/10.1016/j.jaad.2015.07.045 pmid: 26470623
16 A Alikhan, PJ Lynch, DB Eisen. Hidradenitis suppurativa: a comprehensive review. J Am Acad Dermatol 2009; 60(4): 539–561, quiz 562–563
https://doi.org/10.1016/j.jaad.2008.11.911 pmid: 19293006
17 RK Andersen, GB Jemec. Treatments for hidradenitis suppurativa. Clin Dermatol 2017; 35(2): 218–224
https://doi.org/10.1016/j.clindermatol.2016.10.018 pmid: 28274363
18 EJ Giamarellos-Bourboulis, A Antonopoulou, C Petropoulou, M Mouktaroudi, E Spyridaki, F Baziaka, A Pelekanou, H Giamarellou, NG Stavrianeas. Altered innate and adaptive immune responses in patients with hidradenitis suppurativa. Br J Dermatol 2007; 156(1): 51–56
https://doi.org/10.1111/j.1365-2133.2006.07556.x pmid: 17199566
19 Q Chen, H Bao, H Wu, S Zhao, S Huang, F Zhao. Diagnosis of cobalamin C deficiency with renal abnormality from onset in a Chinese child by next generation sequencing: a case report. Exp Ther Med 2017; 14(4): 3637–3643
https://doi.org/10.3892/etm.2017.4970 pmid: 29042959
20 TP Sullivan, E Welsh, FA Kerdel, AE Burdick, RS Kirsner. Infliximab for hidradenitis suppurativa. Br J Dermatol 2003; 149(5): 1046–1049
https://doi.org/10.1111/j.1365-2133.2003.05663.x pmid: 14632813
21 A Kurayev, H Ashkar, A Saraiya, AB Gottlieb. Hidradenitis suppurativa: review of the pathogenesis and treatment. J Drugs Dermatol 2016; 15(8): 1017–1022
pmid: 27538005
22 G Kelly, R Hughes, T McGarry, M van den Born, K Adamzik, R Fitzgerald, C Lawlor, AM Tobin, CM Sweeney, B Kirby. Dysregulated cytokine expression in lesional and nonlesional skin in hidradenitis suppurativa. Br J Dermatol 2015; 173(6): 1431–1439
https://doi.org/10.1111/bjd.14075 pmid: 26282467
23 L Tong, RM Corrales, Z Chen, AL Villarreal, CS De Paiva, R Beuerman, DQ Li, SC Pflugfelder. Expression and regulation of cornified envelope proteins in human corneal epithelium. Invest Ophthalmol Vis Sci 2006; 47(5): 1938–1946
https://doi.org/10.1167/iovs.05-1129 pmid: 16639001
24 Affymetrix. GeneChip® Expression Analysis. Data Analysis Fundamentals. 2004
25 S Anders, W Huber. Differential expression of RNA-Seq data at the gene level — the DESeq package. DESeq version 1.38.0. European Molecular Biology Laboratory (EMBL). 2013
26 L Wang, Z Feng, X Wang, X Wang, X Zhang. DEGseq: an R package for identifying differentially expressed genes from RNA-seq data. Bioinformatics 2010; 26(1): 136–138
https://doi.org/10.1093/bioinformatics/btp612 pmid: 19855105
27 Y Nomura, T Nomura, S Suzuki, M Takeda, O Mizuno, Y Ohguchi, R Abe, Y Murata, H Shimizu. A novel NCSTN mutation alone may be insufficient for the development of familial hidradenitis suppurativa. J Dermatol Sci 2014; 74(2): 180–182
https://doi.org/10.1016/j.jdermsci.2014.01.013 pmid: 24581508
28 X Zhang, SS Sisodia. Acne inversa caused by missense mutations in NCSTN is not fully compatible with impairments in Notch signaling. J Invest Dermatol 2015; 135(2): 618–620
https://doi.org/10.1038/jid.2014.399 pmid: 25211177
29 X Xiao, Y He, C Li, X Zhang, H Xu, B Wang. Nicastrin mutations in familial acne inversa impact keratinocyte proliferation and differentiation through the Notch and phosphoinositide 3-kinase/AKT signalling pathways. Br J Dermatol 2016; 174(3): 522–532
https://doi.org/10.1111/bjd.14223 pmid: 26473517
30 L Yang, C Mao, Y Teng, W Li, J Zhang, X Cheng, X Li, X Han, Z Xia, H Deng, X Yang. Targeted disruption of Smad4 in mouse epidermis results in failure of hair follicle cycling and formation of skin tumors. Cancer Res 2005; 65(19): 8671–8678
https://doi.org/10.1158/0008-5472.CAN-05-0800 pmid: 16204035
31 A Ramirez, A Page, A Gandarillas, J Zanet, S Pibre, M Vidal, L Tusell, A Genesca, DA Whitaker, DW Melton, JL Jorcano. A keratin K5Cre transgenic line appropriate for tissue-specific or generalized Cre-mediated recombination. Genesis 2004; 39(1): 52–57
https://doi.org/10.1002/gene.20025 pmid: 15124227
32 B Wang, W Yang, W Wen, J Sun, B Su, B Liu, D Ma, D Lv, Y Wen, T Qu, M Chen, M Sun, Y Shen, X Zhang. γ-Secretase gene mutations in familial acne inversa. Science 2010; 330(6007): 1065
https://doi.org/10.1126/science.1196284 pmid: 20929727
33 AE Pink, MA Simpson, N Desai, RC Trembath, JNW Barker. γ-Secretase mutations in hidradenitis suppurativa: new insights into disease pathogenesis. J Invest Dermatol 2013; 133(3): 601–607
https://doi.org/10.1038/jid.2012.372 pmid: 23096707
34 MA Boutet, G Bart, M Penhoat, J Amiaud, B Brulin, C Charrier, F Morel, JC Lecron, M Rolli-Derkinderen, A Bourreille, S Vigne, C Gabay, G Palmer, B Le Goff, F Blanchard. Distinct expression of interleukin (IL)-36a, b and g, their antagonist IL-36Ra and IL-38 in psoriasis, rheumatoid arthritis and Crohn’s disease. Clin Exp Immunol 2016; 184(2): 159–173
https://doi.org/10.1111/cei.12761 pmid: 26701127
35 AE Pink, MA Simpson, GW Brice, CH Smith, N Desai, PS Mortimer, JN Barker, RC Trembath. PSENEN and NCSTN mutations in familial hidradenitis suppurativa (acne inversa). J Invest Dermatol 2011; 131(7): 1568–1570
https://doi.org/10.1038/jid.2011.42 pmid: 21412258
36 H Xu, X Xiao, Y Hui, X Zhang, Y He, C Li, B Wang. Phenotype of 53 Chinese individuals with nicastrin gene mutations in association with familial hidradenitis suppurativa (acne inversa). Br J Dermatol 2016; 174(4): 927–929
https://doi.org/10.1111/bjd.14268 pmid: 26522179
37 HH van der Zee, JD Laman, J Boer, EP Prens. Hidradenitis suppurativa: viewpoint on clinical phenotyping, pathogenesis and novel treatments. Exp Dermatol 2012; 21(10): 735–739
https://doi.org/10.1111/j.1600-0625.2012.01552.x pmid: 22882284
38 JW van der Meer, A Simon. The challenge of autoinflammatory syndromes: with an emphasis on hyper-IgD syndrome. Rheumatology (Oxford) 2016; 55(suppl 2): ii23–ii29
https://doi.org/10.1093/rheumatology/kew351 pmid: 27856657
39 S Hessam, M Sand, T Gambichler, M Skrygan, I Rüddel, FG Bechara. Interleukin-36 in hidradenitis suppurativa: evidence for a distinctive proinflammatory role and a key factor in the development of an inflammatory loop. Br J Dermatol 2018; 178(3): 761–767
https://doi.org/10.1111/bjd.16019 pmid: 28975626
40 E Witte-Händel, K Wolk, A Tsaousi, ML Irmer, R Mößner, O Shomroni, T Lingner, K Witte, D Kunkel, G Salinas, S Jodl, N Schmidt, W Sterry, HD Volk, EJ Giamarellos-Bourboulis, A Pokrywka, WD Döcke, S Schneider-Burrus, R Sabat. The IL-1 pathway is hyperactive in hidradenitis suppurativa and contributes to skin infiltration and destruction. J Invest Dermatol 2019; 139(6): 1294–1305
https://doi.org/10.1016/j.jid.2018.11.018 pmid: 30528824
41 R Thomi, M Kakeda, N Yawalkar C Schlapbach, RE Hunger. Increased expression of the interleukin-36 cytokines in lesions of hidradenitis suppurativa. J Eur Acad Dermatol Venereol 2017; 31(12): 2091–2096
https://doi.org/10.1111/jdv.14389
42 R Di Caprio, A Balato, G Caiazzo, S Lembo, A Raimondo, G Fabbrocini, G Monfrecola. IL-36 cytokines are increased in acne and hidradenitis suppurativa. Arch Dermatol Res 2017; 309(8): 673–678
https://doi.org/10.1007/s00403-017-1769-5 pmid: 28852851
43 DE Smith, BR Renshaw, RR Ketchem, M Kubin, KE Garka, JE Sims. Four new members expand the interleukin-1 superfamily. J Biol Chem 2000; 275(2): 1169–1175
https://doi.org/10.1074/jbc.275.2.1169 pmid: 10625660
44 Y Carrier, HL Ma, HE Ramon, L Napierata, C Small, M O’Toole, DA Young, LA Fouser, C Nickerson-Nutter, M Collins, K Dunussi-Joannopoulos, QG Medley. Inter-regulation of Th17 cytokines and the IL-36 cytokines in vitro and in vivo: implications in psoriasis pathogenesis. J Invest Dermatol 2011; 131(12): 2428–2437
https://doi.org/10.1038/jid.2011.234 pmid: 21881584
45 H Blumberg, H Dinh, ES Trueblood, J Pretorius, D Kugler, N Weng, ST Kanaly, JE Towne, CR Willis, MK Kuechle, JE Sims, JJ Peschon. Opposing activities of two novel members of the IL-1 ligand family regulate skin inflammation. J Exp Med 2007; 204(11): 2603–2614
https://doi.org/10.1084/jem.20070157 pmid: 17908936
46 JE Towne, KE Garka, BR Renshaw, GD Virca, JE Sims. Interleukin (IL)-1F6, IL-1F8, and IL-1F9 signal through IL-1Rrp2 and IL-1RAcP to activate the pathway leading to NF-κB and MAPKs. J Biol Chem 2004; 279(14): 13677–13688
https://doi.org/10.1074/jbc.M400117200 pmid: 14734551
47 S Vigne, G Palmer, P Martin, C Lamacchia, D Strebel, E Rodriguez, ML Olleros, D Vesin, I Garcia, F Ronchi, F Sallusto, JE Sims, C Gabay. IL-36 signaling amplifies Th1 responses by enhancing proliferation and Th1 polarization of naive CD4+ T cells. Blood 2012; 120(17): 3478–3487
https://doi.org/10.1182/blood-2012-06-439026 pmid: 22968459
48 BC Melnik, G Plewig. Impaired Notch-MKP-1 signalling in hidradenitis suppurativa: an approach to pathogenesis by evidence from translational biology. Exp Dermatol 2013; 22(3): 172–177
https://doi.org/10.1111/exd.12098 pmid: 23489419
49 K Wolk, J Wenzel, A Tsaousi, E Witte-Händel, N Babel, C Zelenak, HD Volk, W Sterry, S Schneider-Burrus, R Sabat. Lipocalin-2 is expressed by activated granulocytes and keratinocytes in affected skin and reflects disease activity in acne inversa/hidradenitis suppurativa. Br J Dermatol 2017; 177(5): 1385–1393
https://doi.org/10.1111/bjd.15424 pmid: 28256718
50 PM Steinert, E Candi, T Kartasova, L Marekov. Small proline-rich proteins are cross-bridging proteins in the cornified cell envelopes of stratified squamous epithelia. J Struct Biol 1998; 122(1-2): 76–85
https://doi.org/10.1006/jsbi.1998.3957 pmid: 9724607
51 N Wakabayashi, S Shin, SL Slocum, ES Agoston, J Wakabayashi, MK Kwak, V Misra, S Biswal, M Yamamoto, TW Kensler. Regulation of notch1 signaling by nrf2: implications for tissue regeneration. Sci Signal 2010; 3(130): ra52
https://doi.org/10.1126/scisignal.2000762 pmid: 20628156
52 M Schfer, H Farwanah, AH Willrodt, AJ Huebner, K Sandhoff, D Roop, D Hohl, W Bloch, S Werner. Nrf2 links epidermal barrier function with antioxidant defense. EMBO Mol Med 2012(4): 364–379
https://doi.org/10.1002/emmm.201200219
53 N Wakabayashi, DV Chartoumpekis, TW Kensler. Crosstalk between Nrf2 and Notch signaling. Free Radic Biol Med 2015; 88(Pt B): 158–167
https://doi.org/10.1016/j.freeradbiomed.2015.05.017 pmid: 26003520
54 N Wakabayashi, JJ Skoko, DV Chartoumpekis, S Kimura, SL Slocum, K Noda, DL Palliyaguru, M Fujimuro, PA Boley, Y Tanaka, N Shigemura, S Biswal, M Yamamoto, TW Kensler. Notch-Nrf2 axis: regulation of Nrf2 gene expression and cytoprotection by Notch signaling. Mol Cell Biol 2014; 34(4): 653–663
https://doi.org/10.1128/MCB.01408-13 pmid: 24298019
[1] FMD-19031-OF-ZX_suppl_1 Download
Viewed
Full text


Abstract

Cited

  Shared   
  Discussed