リケラボ論文検索は、全国の大学リポジトリにある学位論文・教授論文を一括検索できる論文検索サービスです。

リケラボ 全国の大学リポジトリにある学位論文・教授論文を一括検索するならリケラボ論文検索大学・研究所にある論文を検索できる

リケラボ 全国の大学リポジトリにある学位論文・教授論文を一括検索するならリケラボ論文検索大学・研究所にある論文を検索できる

大学・研究所にある論文を検索できる 「Trapping of CDC42 C-terminal variants in the Golgi drives pyrin inflammasome hyperactivation」の論文概要。リケラボ論文検索は、全国の大学リポジトリにある学位論文・教授論文を一括検索できる論文検索サービスです。

コピーが完了しました

URLをコピーしました

論文の公開元へ論文の公開元へ
書き出し

Trapping of CDC42 C-terminal variants in the Golgi drives pyrin inflammasome hyperactivation

Isa, Masahiko 京都大学 DOI:10.14989/doctor.k24500

2023.03.23

概要

Autoinflammatory disorders are caused by dysregulated activation of innate immune systems and typically present in early
childhood with fever and disease-specific patterns of organ inflammation. The concept of autoinflammatory diseases was proposed in 1999, and since that time a growing number of genetic
causes have been identified for a variety of diseases (Manthiram
et al., 2017; McDermott et al., 1999). Disease-based research has
revealed the molecular mechanisms of excessive innate immune
responses that drive autoinflammatory phenotypes, and these discoveries have provided us with novel therapeutic targets that might
be used to effectively treat these conditions (Nigrovic et al., 2020). ...

この論文で使われている画像

参考文献

Downloaded from http://rupress.org/jem/article-pdf/219/6/e20211889/1432512/jem_20211889.pdf by Kyoto University Igaku Toshokan user on 30 April 2022

Nucleofection and flow cytometry

THP-1 nucleofection and cell death assays were performed as

described previously (Fujisawa et al., 2007; Honda et al., 2021)

with some modifications. THP-1 cells (1 × 106) were transfected

with 500 ng plasmid encoding pAcGFP-fused CDC42 variants or

hMEFV-pAcGFP1-t2a-CDC42 using 4D-Nucleofector and the SG

Cell Line 4D-Nucleofector X Kit (Lonza). Immediately after nucleofection, 10 ng/ml phorbol 12-myristate 13-acetate (FUJIFILM

Wako) was added and the cells were cultured overnight. The

next day, the cells were stained with LIVE/DEAD Fixable Violet

Dead Cell Stain Kit (Thermo Fisher Scientific) as indicated. Cells

were analyzed using a FACSVerse flow cytometer (Becton and

Dickinson Bioscience [BD]) and FlowJo software (BD). Cell death

was calculated as the percentage of the size-gated cells that were

BV421-high.

Bekhouche, B., A. Tourville, Y. Ravichandran, R. Tacine, L. Abrami, M.

Dussiot, A. Khau-Dancasius, O. Boccara, M. Khirat, M. Mangeney, et al.

2020. A toxic palmitoylation of Cdc42 enhances NF-κB signaling and

drives a severe autoinflammatory syndrome. J. Allergy Clin. Immunol.

146:1201–1204.e8. https://doi.org/10.1016/j.jaci.2020.03.020

Bryceson, Y.T., D. Pende, A. Maul-Pavicic, K.C. Gilmour, H. Ufheil, T. Vraetz,

S.C. Chiang, S. Marcenaro, R. Meazza, I. Bondzio, et al. 2012. A prospective evaluation of degranulation assays in the rapid diagnosis of

familial hemophagocytic syndromes. Blood. 119:2754–2763. https://doi

.org/10.1182/blood-2011-08-374199

Bucciol, G., B. Pillay, J. Casas-Martin, S. Delafontaine, M. Proesmans, N.

Lorent, J. Coolen, T. Tousseyn, X. Bossuyt, C.S. Ma, et al. 2020. Systemic

inflammation and myelofibrosis in a patient with takenouchi-kosaki

Journal of Experimental Medicine

https://doi.org/10.1084/jem.20211889

13 of 15

Nishitani-Isa et al.

Pyrin overactivation by CDC42 C-terminal variants

macrophage activation syndrome: A systematic review. Clin. Exp. Immunol. 203:174–182. https://doi.org/10.1111/cei.13543

Lam, M.T., S. Coppola, O.H.F. Krumbach, G. Prencipe, A. Insalaco, C. Cifaldi, I.

Brigida, E. Zara, S. Scala, S. Di Cesare, et al. 2019. A novel disorder

involving dyshematopoiesis, inflammation, and HLH due to aberrant

CDC42 function. J. Exp. Med. 216:2778–2799. https://doi.org/10.1084/jem

.20190147

Li, H.L., N. Fujimoto, N. Sasakawa, S. Shirai, T. Ohkame, T. Sakuma, M.

Tanaka, N. Amano, A. Watanabe, H. Sakurai, et al. 2015. Precise correction of the dystrophin gene in duchenne muscular dystrophy patient

induced pluripotent stem cells by TALEN and CRISPR-Cas9. Stem Cell

Rep. 4:143–154. https://doi.org/10.1016/j.stemcr.2014.10.013

Li, H.L., P. Gee, K. Ishida, and A. Hotta. 2016. Efficient genomic correction

methods in human iPS cells using CRISPR-Cas9 system. Methods. 101:

27–35. https://doi.org/10.1016/j.ymeth.2015.10.015

Magnotti, F., L. Lefeuvre, S. Benezech, T. Malsot, L. Waeckel, A. Martin, S.

Kerever, D. Chirita, M. Desjonqueres, A. Duquesne, et al. 2019. Pyrin

dephosphorylation is sufficient to trigger inflammasome activation in

familial Mediterranean fever patients. EMBO Mol Med. 11:e10547.

https://doi.org/10.15252/emmm.201910547

Manthiram, K., Q. Zhou, I. Aksentijevich, and D.L. Kastner. 2017. The monogenic autoinflammatory diseases define new pathways in human innate immunity and inflammation. Nat. Immunol. 18:832–842. https://doi

.org/10.1038/ni.3777

Martinelli, S., O.H.F. Krumbach, F. Pantaleoni, S. Coppola, E. Amin, L. Pannone, K. Nouri, L. Farina, R. Dvorsky, F. Lepri, et al. 2018. Functional

dysregulation of CDC42 causes diverse developmental phenotypes. Am.

J. Hum. Genet. 102:309–320. https://doi.org/10.1016/j.ajhg.2017.12.015

Masters, S.L., V. Lagou, I. J´eru, P.J. Baker, L. Van Eyck, D.A. Parry, D. Lawless,

D. De Nardo, J.E. Garcia-Perez, L.F. Dagley, et al. 2016. Familial autoinflammation with neutrophilic dermatosis reveals a regulatory

mechanism of pyrin activation. Sci. Transl. Med. 8:332ra45. https://

doi.org/10.1126/scitranslmed.aaf1471

McDermott, M.F., I. Aksentijevich, J. Galon, E.M. McDermott, B.W. Ogunkolade, M. Centola, E. Mansfield, M. Gadina, L. Karenko, T. Pettersson,

et al. 1999. Germline mutations in the extracellular domains of the

55 kDa TNF receptor, TNFR1, define a family of dominantly inherited

autoinflammatory syndromes. Cell. 97:133–144. https://doi.org/10.1016/

s0092-8674(00)80721-7

Moorman, J.P., D. Luu, J. Wickham, D.A. Bobak, and C.S. Hahn. 1999. A balance of signaling by Rho family small GTPases RhoA, Rac1 and Cdc42

coordinates cytoskeletal morphology but not cell survival. Oncogene. 18:

47–57. https://doi.org/10.1038/sj.onc.1202262

Mukai, K., E. Ogawa, R. Uematsu, Y. Kuchitsu, F. Kiku, T. Uemura, S. Waguri,

T. Suzuki, N. Dohmae, H. Arai, et al. 2021. Homeostatic regulation of

STING by retrograde membrane traffic to the ER. Nat. Commun. 12:61.

https://doi.org/10.1038/s41467-020-20234-9

Nagai, K., K. Yamamoto, H. Fujiwara, J. An, T. Ochi, K. Suemori, T. Yasumi, H.

Tauchi, K. Koh, M. Sato, et al. 2010. Subtypes of familial hemophagocytic lymphohistiocytosis in Japan based on genetic and functional

analyses of cytotoxic T lymphocytes. PLoS One. 5:e14173. https://doi.org/

10.1371/journal.pone.0014173

Nakagawa, M., Y. Taniguchi, S. Senda, N. Takizawa, T. Ichisaka, K. Asano, A.

Morizane, D. Doi, J. Takahashi, M. Nishizawa, et al. 2014. A novel efficient feeder-free culture system for the derivation of human induced

pluripotent stem cells. Sci. Rep. 4:3594. https://doi.org/10.1038/

srep03594

Nigrovic, P.A., P.Y. Lee, and H.M. Hoffman. 2020. Monogenic autoinflammatory disorders: Conceptual overview, phenotype, and clinical

approach. J. Allergy Clin. Immunol. 146:925–937. https://doi.org/10.1016/j

.jaci.2020.08.017

Park, Y.H., G. Wood, D.L. Kastner, and J.J. Chae. 2016. Pyrin inflammasome

activation and RhoA signaling in the autoinflammatory diseases FMF

and HIDS. Nat. Immunol. 17:914–921. https://doi.org/10.1038/ni.3457

Saito, M.K. 2021. Elucidation of the pathogenesis of autoinflammatory

diseases using iPS cells. Children. 8:94. https://doi.org/10.3390/

children8020094

Shibata, H., T. Yasumi, S. Shimodera, E. Hiejima, K. Izawa, T. Kawai, R.

Shirakawa, T. Wada, R. Nishikomori, H. Horiuchi, et al. 2018. Human

CTL-based functional analysis shows the reliability of a munc13-4

protein expression assay for FHL3 diagnosis. Blood. 131:2016–2025.

https://doi.org/10.1182/blood-2017-10-812503

Szczawinska-Poplonyk, A., R. Ploski, E. Bernatowska, and M. Pac. 2020. A novel

CDC42 mutation in an 11-year old child manifesting as syndromic immunodeficiency, autoinflammation, hemophagocytic lymphohistiocytosis,

Journal of Experimental Medicine

https://doi.org/10.1084/jem.20211889

Downloaded from http://rupress.org/jem/article-pdf/219/6/e20211889/1432512/jem_20211889.pdf by Kyoto University Igaku Toshokan user on 30 April 2022

syndrome due to CDC42 Tyr64Cys mutation. J. Clin. Immunol. 40:

567–570. https://doi.org/10.1007/s10875-020-00742-5

Chae, J.J., Y.H. Cho, G.S. Lee, J. Cheng, P.P. Liu, L. Feigenbaum, S.I. Katz,

and D.L. Kastner. 2011. Gain-of-function Pyrin mutations induce

NLRP3 protein-independent interleukin-1β activation and severe

autoinflammation in mice. Immunity. 34:755–768. https://doi.org/

10.1016/j.immuni.2011.02.020

Etienne-Manneville, S. 2004. Cdc42-the centre of polarity. J. Cell Sci. 117:

1291–1300. https://doi.org/10.1242/jcs.01115

Fu, Y., J.A. Foden, C. Khayter, M.L. Maeder, D. Reyon, J.K. Joung, and J.D.

Sander. 2013. High-frequency off-target mutagenesis induced by

CRISPR-Cas nucleases in human cells. Nat. Biotechnol. 31:822–826.

https://doi.org/10.1038/nbt.2623

Fujisawa, A., N. Kambe, M. Saito, R. Nishikomori, H. Tanizaki, N. Kanazawa,

S. Adachi, T. Heike, J. Sagara, T. Suda, et al. 2007. Disease-associated

mutations in CIAS1 induce cathepsin B-dependent rapid cell death of

human THP-1 monocytic cells. Blood. 109:2903–2911. https://doi.org/10

.1182/blood-2006-07-033597

Gao, W., J. Yang, W. Liu, Y. Wang, and F. Shao. 2016. Site-specific phosphorylation and microtubule dynamics control Pyrin inflammasome

activation. Proc. Natl. Acad. Sci. USA. 113:E4857–E4866. https://doi.org/

10.1073/pnas.1601700113

Garcia-Mata, R., E. Boulter, and K. Burridge. 2011. The “invisible hand”:

Regulation of RHO GTPases by RHOGDIs. Nat. Rev. Mol. Cell Biol. 12:

493–504. https://doi.org/10.1038/nrm3153

Gaudelli, N.M., A.C. Komor, H.A. Rees, M.S. Packer, A.H. Badran, D.I. Bryson,

and D.R. Liu. 2017. Programmable base editing of A•T to G•C in genomic DNA without DNA cleavage. Nature. 551:464–471. https://doi.org/

10.1038/nature24644

Gernez, Y., A.A. De Jesus, H. Alsaleem, C. Macaubas, A. Roy, D. Lovell, K.A.

Jagadeesh, S. Alehashemi, L. Erdman, M. Grimley, et al. 2019. Severe

autoinflammation in 4 patients with C-terminal variants in cell division

control protein 42 homolog (CDC42) successfully treated with IL-1β inhibition. J. Allergy Clin. Immunol. 144:1122–1125.e6. https://doi.org/10

.1016/j.jaci.2019.06.017

Gibson, R.M., and A.L. Wilson-Delfosse. 2001. RhoGDI-binding-defective

mutant of Cdc42Hs targets to membranes and activates filopodia formation but does not cycle with the cytosol of mammalian cells. Biochem.

J. 359:285–294. https://doi.org/10.1042/0264-6021:3590285

Heasman, S.J., and A.J. Ridley. 2008. Mammalian Rho GTPases: New insights

into their functions from in vivo studies. Nat. Rev. Mol. Cell Biol. 9:

690–701. https://doi.org/10.1038/nrm2476

Honda, Y., Y. Maeda, K. Izawa, T. Shiba, T. Tanaka, H. Nakaseko, K. Nishimura, H. Mukoyama, M. Isa-Nishitani, T. Miyamoto, et al. 2021. Rapid

flow cytometry-based assay for the functional classification of MEFV

variants. J. Clin. Immunol. 41:1187–1197. https://doi.org/10.1007/s10875

-021-01021-7

Hori, M., T. Yasumi, S. Shimodera, H. Shibata, E. Hiejima, H. Oda, K. Izawa, T.

Kawai, M. Ishimura, N. Nakano, et al. 2017. A CD57+ CTL degranulation

assay effectively identifies familial hemophagocytic lymphohistiocytosis type 3 patients. J. Clin. Immunol. 37:92–99. https://doi.org/10.1007/

s10875-016-0357-3

Huang, T.P., K.T. Zhao, S.M. Miller, N.M. Gaudelli, B.L. Oakes, C. Fellmann,

D.F. Savage, and D.R. Liu. 2019. Circularly permuted and PAM-modified

Cas9 variants broaden the targeting scope of base editors. Nat. Biotechnol. 37:626–631. https://doi.org/10.1038/s41587-019-0134-y

Kang, R., J. Wan, P. Arstikaitis, H. Takahashi, K. Huang, A.O. Bailey, J.X.

Thompson, A.F. Roth, R.C. Drisdel, R. Mastro, et al. 2008. Neural

palmitoyl-proteomics reveals dynamic synaptic palmitoylation. Nature.

456:904–909. https://doi.org/10.1038/nature07605

Kawasaki, Y., H. Oda, J. Ito, A. Niwa, T. Tanaka, A. Hijikata, R. Seki, A. Nagahashi, M. Osawa, I. Asaka, et al. 2017. Identification of a highfrequency somatic NLRC4 mutation as a cause of autoinflammation

by pluripotent cell-based phenotype dissection. Arthritis Rheumatol. 69:

447–459. https://doi.org/10.1002/art.39960

Koblan, L.W., J.L. Doman, C. Wilson, J.M. Levy, T. Tay, G.A. Newby, J.P.

Maianti, A. Raguram, and D.R. Liu. 2018. Improving cytidine and adenine base editors by expression optimization and ancestral reconstruction. Nat. Biotechnol. 36:843–846. https://doi.org/10.1038/nbt.4172

Komor, A.C., Y.B. Kim, M.S. Packer, J.A. Zuris, and D.R. Liu. 2016. Programmable editing of a target base in genomic DNA without doublestranded DNA cleavage. Nature. 533:420–424. https://doi.org/10.1038/

nature17946

Krei, J.M., H.J. Møller, and J.B. Larsen. 2021. The role of interleukin-18 in the

diagnosis and monitoring of hemophagocytic lymphohistiocytosis/

14 of 15

Nishitani-Isa et al.

Pyrin overactivation by CDC42 C-terminal variants

Wan, J., A.F. Roth, A.O. Bailey, and N.G. Davis. 2007. Palmitoylated proteins:

Purification and identification. Nat. Protoc. 2:1573–1584. https://doi.org/

10.1038/nprot.2007.225

Watkin, L.B., B. Jessen, W. Wiszniewski, T.J. Vece, M. Jan, Y. Sha, M. Thamsen,

R.L.P. Santos-Cortez, K. Lee, T. Gambin, et al. 2015. COPA mutations impair

ER-Golgi transport and cause hereditary autoimmune-mediated lung disease

and arthritis. Nat. Genet. 47:654–660. https://doi.org/10.1038/ng.3279

Weiss, E.S., C. Girard-Guyonvarc’h, D. Holzinger, A.A. de Jesus, Z. Tariq, J.

Picarsic, E.J. Schiffrin, D. Foell, A.A. Grom, S. Ammann, et al. 2018.

Interleukin-18 diagnostically distinguishes and pathogenically promotes human and murine macrophage activation syndrome. Blood. 131:

1442–1455. https://doi.org/10.1182/blood-2017-12-820852

Wilson, A.L., R.A. Erdman, F. Castellano, and W.A. Maltese. 1998. Prenylation

of Rab8 GTPase by type I and type II geranylgeranyl transferases. Biochem. J. 333:497–504. https://doi.org/10.1042/bj3330497

Wu, W.J., J.W. Erickson, R. Lin, and R.A. Cerione. 2000. The gamma-subunit

of the coatomer complex binds Cdc42 to mediate transformation. Nature. 405:800–804. https://doi.org/10.1038/35015585

Journal of Experimental Medicine

https://doi.org/10.1084/jem.20211889

Downloaded from http://rupress.org/jem/article-pdf/219/6/e20211889/1432512/jem_20211889.pdf by Kyoto University Igaku Toshokan user on 30 April 2022

and malignancy: A case report. Front. Immunol. 11:318. https://doi.org/10

.3389/fimmu.2020.00318

Takenouchi, T., R. Kosaki, T. Niizuma, K. Hata, and K. Kosaki. 2015. Macrothrombocytopenia and developmental delay with a de novo CDC42

mutation: Yet another locus for thrombocytopenia and developmental

delay. Am. J. Med. Genet. A. 167A:2822–2825. https://doi.org/10.1002/

ajmg.a.37275

Takenouchi, T., N. Okamoto, S. Ida, T. Uehara, and K. Kosaki. 2016. Further

evidence of a mutation in CDC42 as a cause of a recognizable syndromic

form of thrombocytopenia. Am. J. Med. Genet. A. 170A:852–855. https://

doi.org/10.1002/ajmg.a.37526

Van Gorp, H., P.H.V. Saavedra, N.M. De Vasconcelos, N. Van Opdenbosch, L. Vande Walle, M. Matusiak, G. Prencipe, A. Insalaco, F. Van Hauwermeiren, D. Demon, et al. 2016. Familial

Mediterranean fever mutations lift the obligatory requirement

for microtubules in Pyrin inflammasome activation. Proc. Natl.

Acad. Sci. USA. 113:14384–14389. https://doi.org/10.1073/pnas

.1613156113

15 of 15

Supplemental material

Downloaded from http://rupress.org/jem/article-pdf/219/6/e20211889/1432512/jem_20211889.pdf by Kyoto University Igaku Toshokan user on 30 April 2022

Nishitani-Isa et al.

Pyrin overactivation by CDC42 C-terminal variants

Journal of Experimental Medicine

https://doi.org/10.1084/jem.20211889

S1

Downloaded from http://rupress.org/jem/article-pdf/219/6/e20211889/1432512/jem_20211889.pdf by Kyoto University Igaku Toshokan user on 30 April 2022

Figure S1. Increased production of IL-1β and IL-18 in patients carrying CDC42R186C. (A) Levels of IL-1β, IL-6, IL-18, and IFNγ in sera of Patients 1 and 2.

(B) IL-18 release in response to various inflammasome stimuli from iPS-derived MPs established from Patient 1 and healthy controls. (C and D) IL-1β and IL-6

release in response to various inflammasome stimuli from iPS-derived MPs (C) and MLs (D) established from Patient 2 and healthy controls. Data are representative of two independent experiments using three independent clones. Statistical significance was determined by Mann–Whitney’s U test. *, P < 0.05.

Nishitani-Isa et al.

Pyrin overactivation by CDC42 C-terminal variants

Journal of Experimental Medicine

https://doi.org/10.1084/jem.20211889

S2

Downloaded from http://rupress.org/jem/article-pdf/219/6/e20211889/1432512/jem_20211889.pdf by Kyoto University Igaku Toshokan user on 30 April 2022

Figure S2. Immunoblot analysis of CDC42 C-terminal variants, cytokine production from iPS-MLs carrying CDC42R66G, and the effect of CDC42 silencing on cytokine production from CDC42WT and CDC42R186C iPS-MLs. (A) CDC42 C-terminal variants were transiently transfected into HEK293T cells,

and their expression levels were evaluated by Western blot analysis. (B) iPSCs carrying CDC42R66G were generated by manipulating WT-iPSCs by single-base

editing and iPS-MPs were differentiated. Cells were stimulated with LPS + TcdA and the production of IL-1β and IL-6 was evaluated. (C) iPS-MLs from Patient

1 and healthy controls were treated with scramble or CDC42 siRNA and the release of IL-1β and IL-6 was monitored in response to LPS + TcdA or LPS + nigericin

stimulation. Immunoblot images at the bottom show the effects of siRNAs. Representative results of (A) two independent experiments with similar results, and

(B and C) two independent experiments with three clones, are shown. Statistical significance was determined by Mann–Whitney’s U test (B) and by Dunnet

test (C). *, P < 0.05; **, P < 0.01; ns, not significant. Source data are available for this figure: SourceData FS2.

Nishitani-Isa et al.

Pyrin overactivation by CDC42 C-terminal variants

Journal of Experimental Medicine

https://doi.org/10.1084/jem.20211889

S3

Downloaded from http://rupress.org/jem/article-pdf/219/6/e20211889/1432512/jem_20211889.pdf by Kyoto University Igaku Toshokan user on 30 April 2022

Figure S3. Analyses of NF-κB signaling in iPS-MLs and cytolytic and degranulation activity of NK cells and CTLs from Patient 1. (A and B) Immunoblot

of JNK, ERK, P65, P38 (A) and pyrin (B) molecules and their phosphorylated forms in iPS-MLs from Patient 1 and a healthy control in response to LPS (A) and

LPS ± TcdA (B) stimuli. (C) Cytolytic activity of activated NK cells derived from healthy controls, Patient 1, and a FHL2 patient. (D and E) Degranulation of

activated NK cells (D) and CTLs (E) derived from healthy controls, Patient 1, and a FHL3 patient. Representative results of three (A and B) and two (C–E)

independent experiments with similar results are shown. FHL, familial hemophagocytic lymphohistiocytosis. Source data are available for this figure: SourceData FS3.

Nishitani-Isa et al.

Pyrin overactivation by CDC42 C-terminal variants

Journal of Experimental Medicine

https://doi.org/10.1084/jem.20211889

S4

Downloaded from http://rupress.org/jem/article-pdf/219/6/e20211889/1432512/jem_20211889.pdf by Kyoto University Igaku Toshokan user on 30 April 2022

Figure S4. CDC42R186C and CDC42*192C*24 variants do not affect the transport and the localization of COP-I. (A) mScarletI-T2A-CDC42 and VSV-G

(tsO45)-GFP were transiently expressed in COS-1 cells. Cells were incubated at 40°C for 16 h to allow VSV-G protein to accumulate in the ER, and then at 32°C

for 30 min. (B) mScarlet-T2A-CDC42 and EGFP-STING were stably expressed in Sting−/− MEFs as indicated. (C–E) GFP-T2A-CDC42 was transiently expressed

in COS-1 cells. Cells were then fixed and stained for GM130 and for β-COP (C), β9-COP (D), and γ-COP (E). Nuclei were stained with DAPI (blue). Representative

results of two independent experiments with similar results are shown. Scale bars, 10 µm.

Nishitani-Isa et al.

Pyrin overactivation by CDC42 C-terminal variants

Journal of Experimental Medicine

https://doi.org/10.1084/jem.20211889

S5

Downloaded from http://rupress.org/jem/article-pdf/219/6/e20211889/1432512/jem_20211889.pdf by Kyoto University Igaku Toshokan user on 30 April 2022

Figure S5. Addition of the C-terminal sequence of CDC42 (R186C) to Rac1 alters its localization to the Golgi. Rac1 or Rac1-CDC42 (C-terminal) was

transiently expressed in COS-1 cells. Cells were then fixed, permeabilized, and stained for GM130. Nuclei were stained with DAPI (blue). The magnified image of

the perinuclear region is shown in the lower left box. Representative results of two independent experiments with similar results are shown. Scale bars, 10 µm.

Nishitani-Isa et al.

Pyrin overactivation by CDC42 C-terminal variants

Journal of Experimental Medicine

https://doi.org/10.1084/jem.20211889

S6

...

参考文献をもっと見る

全国の大学の
卒論・修論・学位論文

一発検索!

この論文の関連論文を見る