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 「BRCA1 haploinsufficiency promotes chromosomal amplification under Fenton reaction-based carcinogenesis through ferroptosis-resistance」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

BRCA1 haploinsufficiency promotes chromosomal amplification under Fenton reaction-based carcinogenesis through ferroptosis-resistance

孔, 颖怡 名古屋大学

2022.12.22

概要

【Introduction】
Germline-mutation in BRCA1 tumor suppressor gene is an established risk for carcinogenesis. However, supporting data is insufficient both in the mutation spectra of cancers in the patients with BRCA1 germline-mutation and in murine models of Brca1 haploinsufficiency. Furthermore, information on the driving force toward carcinogenesis in BRCA1 mutation carriers is lacking. Here we applied Fenton reaction-based renal carcinogenesis to a rat heterozygously knockout model of BRCA1 haploinsufficiency (MUT model; L63X/+). It revealed significant promotion of renal cell carcinoma which identified significant increase in chromosomal amplification, including c-Myc. Subacute-phase analysis in the MUT model revealed dysregulated iron metabolism with mitochondrial malfunction, leading to renal tubular proliferation with iron overload. In conclusion, we demonstrate that biallelic BRCA1 provides protection for mitochondrial metabolism under iron-catalyzed oxidative stress, preventing the emergence of neoplastic cells with chromosomal amplification, which can be a target for cancer prevention and therapeutics.

【Methods】
Rat BRCA1 haploinsufficiency model A rat Brca1-MUT(L63X/+) model was established by Prof. Imaoka, and used in the present study. Rats with homozygously MUT alleles (L63X/L63X) were embryonic lethal. Genotyping was performed with PCR.

Renal carcinogenesis experiments with Fe-NTA Male rats (wild-type and Brca1-MUT) were injected ip with Fe-NTA with a dose of 5~10 mg iron/kg for a total of 11 weeks. The rats which were found to have fatal renal cell carcinoma (RCC) by palpation or to be dying were euthanized. Computed tomography and magnetic resonance imaging were used, if necessary, to confirm the presence of RCC. Hematoxylin & eosin staining and immunohistochemistry were performed.

Study of chromosomal amplification Array-based comparative genomic hybridization (aCGH) was performed with genomic DNA from RCC samples (4/4 in wild-type with/without metastasis, 4/4 in Brca1-MUT with/without metastasis) and analyzed. Fluorescent in situ hybridization (FISH) was performed on FFPE RCC sections using cMyc(Cy3)/Ch7CEN(Spectrum Green) dual color FISH probe.

Subacute study Male rats (wild-type and Brca1-MUT) were injected ip with Fe-NTA for carcinogenesisprotocol either for 1 or 3 week[s]. The rats were euthanized 48h after the final injection. Microarray analyses were performed with total RNA from kidney cortex obtained, which were subsequently analyzed (GEO accession: GSE198507). GO term analysis was performed between wild-type and Brca1-MUT groups. Transmission electron microscopy (TEM) was applied to renal cortical area excised from control and Fe-NTA treated rats of wild-type and Brca1-MUT groups. Immunoblot of Drp1, mTOR, and p62 was examined. Immunohistochemical analysis of Transferrin and Perl’s iron staining were performed to evaluate iron metabolism. Immunoblot of Transferrin, Transferrin Receptor, and Ferritin heavy chain was subsequently applied. To evaluate nuclear mutagenic and ferroptosis environment, immunohistochemistry of 8- hydroxy-2’-deoxyguanosine (8-OHdG; N45.1), Ki-67, -H2AX, and 4-hydroxy-2-nonenalmodified proteins (HNEJ-1) was performed.

【Results】
Brca1 haploinsufficiency significantly promotes Fe-NTA-induced renal carcinogenesis There was a significant promotional effect of renal carcinogenesis in Brca1 haploinsufficiency in comparison to WT (Fig. 1A). RCCs of MUT rats showed a higher but statistically not significant incidence of peritoneal invasion/dissemination (35.3% and 52.9%, P = 0.300; Table 1) with higher Ki-67 cellular proliferation index.

Brca1 haploinsufficiency significantly increases chromosomal amplification in Fe-NTAinduced RCCs We observed that in comparison to WT, MUT RCCs with/without pulmonary metastasis revealed significantly higher frequency of chromosomal amplification, among which amplification of chromosome 7 was most prominent (Fig. 2A). In WT RCCs, deletions were significantly prevalent in general but only centromeric portion of chromosome 4 showed significantly frequent amplification, where c-Met oncogene was located as we previously described (Fig.2A).

Specific amplification of c-Myc oncogene in the RCCs of Brca1 MUT rats We found c-Myc amplification in 4/8 MUT RCCs examined by aCGH whereas only 1/8 of WT RCCs showed it. FISH analysis confirmed significantly higher incidence of c-Myc amplification in the other MUT RCCs in comparison to WT and revealed that c-Myc amplification included those in the extrachromosomal DNA (micronuclei) (Fig. 2B).

Brca1 haploinsufficiency causes iron dysregulation in association with mitochondrial malfunction in the subacute phase of Fe-NTA-induced renal carcinogenesis Microarray analysis between WT and MUT at 3 weeks of carcinogenesis protocol suggested the pathways involved in heme/hemoglobin, oxygen and iron (Fig. 3A), leading to investigation of mitochondria and iron metabolism. We performed TEM analysis, which disclosed that after 3 weeks of Fe-NTA administration, mitochondria became significantly irregular and smallsized with electron-dense deposits and loss of cristae whereas higher lysosomal/autophagosomal fraction was maintained in the Brca1-MUT kidney (Fig. 4A-C). This was confirmed by decrease in Drp1, mTOR, and p62 (Figs. 4D). We found significantly higher iron deposition in the MUT kidney at 3 weeks of carcinogenesis protocol than WT with Perl’s iron staining and Tf expression (Fig. 5AB), which was consistent with increase in Fth1 and decrease in Tfr1(Fig. 5C). Increase in Tf production in the MUT kidney at 3 weeks of carcinogenesis protocol suggests an establishment of a regulatory system to avoid excess iron.

Brca1 haploinsufficiency generates carcinogenic environments with ferroptosis-resistance under iron-catalyzed persistent oxidative stress At 3 weeks of carcinogenesis protocol, 8-OHdG was significantly increased in the MUT kidney with higher Ki-67 cellular proliferation index in comparison to WT (Fig. 6AB). - H2AX, a marker for DNA double-strand breaks, significantly increased in the kidney with WT significantly higher than MUT (Fig. 6C), which may be attributed to Brca1 haploinsufficiency. Immunostaining by HNEJ-1, a novel maker of ferroptosis, showed higher signal in WT than MUT (Fig. 6D), suggesting that MUT kidney is more resistant to ferroptosis under the carcinogenesis protocol.

【Discussion】
Here we demonstrated that Brca1 haploinsufficiency significantly promotes Fe-NTAinduced renal carcinogenesis, indicating the usefulness of this rat MUT model to clarify the responsible molecular mechanisms in comparison to previously failed murine models. We found that c-Myc in chromosome 7 is amplified with a significantly higher probability in the RCCs in Brca1-MUT rats. Therefore, BRCA1 prevents or delays extrachromosomal oncogene amplification under oxidative stress. We believe that whole genome sequencing in the future with other studies would clarify why RCCs in the MUT rats show more aggressive behavior. We found that iron metabolism is significantly altered in the renal tubules in response to persistent Fe-NTA-induced oxidative stress. Tf overexpression appears a strategy of tubular cells to regulate intracellular iron concentration either by releasing or preventing uptake of iron to escape ferroptosis, which needs further investigation. After 3 weeks of Fe-NTA administration, ferroptosis markers were significantly lower in the MUT model indicating ferroptosis-resistance. Further microarray data suggested mitochondrial malfunction by the accumulation of iron- and heme-associated genes. Electron microscopic analysis revealed that deformed mitochondria in the MUT kidney was significantly aggravated after 3-week Fe-NTA administration with smaller mitochondrial mass. Because mitochondria are a central iron metabolism organelle, we assume that mitochondrial dysfunction would dramatically alter the iron distribution intracellularly with vice versa and promote the energy acquisition system to be more dependent on glycolytic system as established in cancer. Finally, we interpret the decrease in -H2AX in Brca1-MUT as an impairment of DNA damage recognition, which needs further investigation. And a decrease in phosphorylated Brca1 may eventually cause failure of cell-cycle checkpoints, leading to genomic amplification.

【Conclusion】
Here we demonstrated that BRCA1 haploinsufficiency causes mitochondrial dysfunction, leading to cellular iron deposition under Fe-NTA-induced renal carcinogenesis model. This can be a carcinogenic driving force to the early establishment of ferroptosis-resistant target cells (Fig. 7). Further, BRCA1 haploinsufficiency facilitates Fe-NTA-induced renal carcinogenesis at the promotional phase and allows more chromosomal amplifications, including c-Myc. Therefore, manipulating the iron metabolism, especially at the target organs, can be a preventive strategy of various carcinogenesis for the BRCA1 germline-mutated patients. Brca1(L63X/+) haploinsufficient rat provides us with a more plausible model to evaluate possible strategies to increase the quality of life of BRCA1 germline-mutated patients.

参考文献

[1] B. Vogelstein, K.W. Kinzler, The Genetic Basis of Human Cancer, McGraw-Hill, New York, 1998.

[2] Y. Miki, J. Swensen, D. Shattuck-Eidens, P.A. Futreal, K. Harshman, S. Tavtigian, Q. Liu, C. Cochran, L.M. Bennett, W. Ding, et al., A strong candidate for the breast and ovarian cancer susceptibility gene BRCA1, Science 266 (5182) (1994) 66–71.

[3] A. Liede, B.Y. Karlan, S.A. Narod, Cancer risks for male carriers of germline mutations in BRCA1 or BRCA2: a review of the literature, J. Clin. Oncol. 22 (4) (2004) 735–742.

[4] H. Cavanagh, K.M. Rogers, The role of BRCA1 and BRCA2 mutations in prostate, pancreatic and stomach cancers, Hered. Cancer Clin. Pract. 13 (1) (2015) 16.

[5] S.A. Narod, W.D. Foulkes, BRCA1 and BRCA2: 1994 and beyond, Nat. Rev. Cancer 4 (9) (2004) 665–676.

[6] N.M. Tung, J.C. Boughey, L.J. Pierce, M.E. Robson, I. Bedrosian, J.R. Dietz, A. Dragun, J.B. Gelpi, E.W. Hofstatter, C.J. Isaacs, I. Jatoi, E. Kennedy, J.K. Litton, N.A. Mayr, R.D. Qamar, M.G. Trombetta, B.E. Harvey, M.R. Somerfield, D. Zakalik, Management of hereditary breast cancer: American society of clinical oncology, American society for radiation oncology, and society of surgical oncology guideline, J. Clin. Oncol. 38 (18) (2020) 2080–2106.

[7] B. Evers, J. Jonkers, Mouse models of BRCA1 and BRCA2 deficiency: past lessons, current understanding and future prospects, Oncogene 25 (43) (2006) 5885–5897.

[8] S. Akatsuka, G.H. Li, S. Toyokuni, Superiority of rat over murine model for studies on the evolution of cancer genome, Free Radic. Res. 52 (11–12) (2018) 1323–1327.

[9] S. Toyokuni, Role of iron in carcinogenesis: cancer as a ferrotoxic disease, Cancer Sci. 100 (1) (2009) 9–16.

[10] S. Toyokuni, F. Ito, K. Yamashita, Y. Okazaki, S. Akatsuka, Iron and thiol redox signaling in cancer: an exquisite balance to escape ferroptosis, Free Radic. Biol. Med. 108 (2017) 610–626.

[11] S.V. Torti, D.H. Manz, B.T. Paul, N. Blanchette-Farra, F.M. Torti, Iron and cancer, Annu. Rev. Nutr. 38 (2018) 97–125.

[12] S. Toyokuni, I. Yanatori, Y. Kong, H. Zheng, Y. Motooka, L. Jiang, Ferroptosis at the crossroads of infection, aging and cancer, Cancer Sci. 111 (2020) 2665–2671.

[13] S. Toyokuni, Y. Kong, Z. Cheng, K. Sato, S. Hayashi, F. Ito, L. Jiang, I. Yanatori, Y. Okazaki, S. Akatsuka, Carcinogenesis as side effects of iron and oxygen utilization: from the unveiled truth toward ultimate bioengineering, Cancers 12 (11) (2020) 3320.

[14] J. Kotsopoulos, G. Sukiennicki, M. Muszynska, D. Gackowski, K. Kaklewski, K. Durda, K. Jaworska, T. Huzarski, J. Gronwald, T. Byrski, O. Ashuryk, T. Debniak, A. Toloczko-Grabarek, M. Stawicka, D. Godlewski, R. Olinski, A. Jakubowska, S. A. Narod, J. Lubinski, Plasma micronutrients, trace elements, and breast cancer in BRCA1 mutation carriers: an exploratory study, Cancer Causes Control 23 (7) (2012) 1065–1074.

[15] S. Toyokuni, K. Uchida, K. Okamoto, Y. Hattori-Nakakuki, H. Hiai, E.R. Stadtman, Formation of 4-hydroxy-2-nonenal-modified proteins in the renal proximal tubules of rats treated with a renal carcinogen, ferric nitrilotriacetate, Proc. Natl. Acad. Sci. U.S.A. 91 (1994) 2616–2620.

[16] S. Toyokuni, T. Mori, M. Dizdaroglu, DNA base modifications in renal chromatin of Wistar rats treated with a renal carcinogen, ferric nitrilotriacetate, Int. J. Cancer 57 (1994) 123–128.

[17] S. Toyokuni, T. Mori, H. Hiai, M. Dizdaroglu, Treatment of Wistar rats with a renal carcinogen, ferric nitrilotriacetate, causes DNA-protein cross-linking between thymine and tyrosine in their renal chromatin, Int. J. Cancer 62 (1995) 309–313.

[18] S. Toyokuni, X.P. Luo, T. Tanaka, K. Uchida, H. Hiai, D.C. Lehotay, Induction of a wide range of C2-12 aldehydes and C7-12 acyloins in the kidney of Wistar rats after treatment with a renal carcinogen, ferric nitrilotriacetate, Free Radic. Biol. Med. 22 (1997) 1019–1027.

[19] Y. Ebina, S. Okada, S. Hamazaki, F. Ogino, J.L. Li, O. Midorikawa, Nephrotoxicity and renal cell carcinoma after use of iron- and aluminum- nitrilotriacetate complexes in rats, J. Natl. Cancer Inst. 76 (1986) 107–113.

[20] Y. Nishiyama, H. Suwa, K. Okamoto, M. Fukumoto, H. Hiai, S. Toyokuni, Low incidence of point mutations in H-, K- and N-ras oncogenes and p53 tumor suppressor gene in renal cell carcinoma and peritoneal mesothelioma of Wistar rats induced by ferric nitrilotriacetate, Jpn. J. Cancer Res. 86 (1995) 1150–1158.

[21] T. Tanaka, Y. Iwasa, S. Kondo, H. Hiai, S. Toyokuni, High incidence of allelic loss on chromosome 5 and inactivation of p15 INK4B and p16 INK4A tumor suppressor genes in oxystress-induced renal cell carcinoma of rats, Oncogene 18 (1999) 3793–3797.

[22] M. Hiroyasu, M. Ozeki, H. Kohda, M. Echizenya, T. Tanaka, H. Hiai, S. Toyokuni, Specific allelic loss of p16 (INK4A) tumor suppressor gene after weeks of ironmediated oxidative damage during rat renal carcinogenesis, Am. J. Pathol. 160 (2) (2002) 419–424.

[23] S. Akatsuka, Y. Yamashita, H. Ohara, Y.T. Liu, M. Izumiya, K. Abe, M. Ochiai, L. Jiang, H. Nagai, Y. Okazaki, H. Murakami, Y. Sekido, E. Arai, Y. Kanai, O. Hino, T. Takahashi, H. Nakagama, S. Toyokuni, Fenton reaction induced cancer in wild type rats recapitulates genomic alterations observed in human cancer, PLoS One 7 (8) (2012), e43403.

[24] S. Toyokuni, The origin and future of oxidative stress pathology: from the recognition of carcinogenesis as an iron addiction with ferroptosisresistance to non-thermal plasma therapy, Pathol. Int. 66 (2016) 245–259.

[25] O.A. Stefansson, J.G. Jonasson, O.T. Johannsson, K. Olafsdottir, M. Steinarsdottir, S. Valgeirsdottir, J.E. Eyfjord, Genomic profiling of breast tumours in relation to BRCA abnormalities and phenotypes, Breast Cancer Res. 11 (4) (2009) R47.

[26] Y. Inagaki-Kawata, K. Yoshida, N. Kawaguchi-Sakita, M. Kawashima, T. Nishimura, N. Senda, Y. Shiozawa, Y. Takeuchi, Y. Inoue, A. Sato-Otsubo, Y. Fujii, Y. Nannya, E. Suzuki, M. Takada, H. Tanaka, Y. Shiraishi, K. Chiba, Y. Kataoka, M. Torii, H. Yoshibayashi, K. Yamagami, R. Okamura, Y. Moriguchi, H. Kato, S. Tsuyuki, A. Yamauchi, H. Suwa, T. Inamoto, S. Miyano, S. Ogawa, M. Toi, Genetic and clinical landscape of breast cancers with germline BRCA1/2 variants, Commun Biol 3 (1) (2020) 578.

[27] S. Toyokuni, S. Okada, S. Hamazaki, M. Fujioka, J.-L. Li, O. Midorikawa, Cirrhosis of the liver induced by cupric nitrilotriacetate in Wistar rats: an experimetnal model of copper toxicosis, Am. J. Pathol. 134 (1989) 1263–1274.

[28] L. Jiang, H. Zheng, Q. Lyu, S. Hayashi, K. Sato, Y. Sekido, K. Nakamura, H. Tanaka, K. Ishikawa, H. Kajiyama, M. Mizuno, M. Hori, S. Toyokuni, Lysosomal nitric oxide determines transition from autophagy to ferroptosis after exposure to plasmaactivated Ringer’s lactate, Redox Biol. 43 (2021) 101989.

[29] H. Zheng, L. Jiang, T. Tsuduki, M. Conrad, S. Toyokuni, Embryonal erythropoiesis and aging exploit ferroptosis, Redox Biol. 48 (2021) 102175.

[30] Z. Cheng, S. Akatsuka, G.H. Li, K. Mori, T. Takahashi, S. Toyokuni, Ferroptosisresistance determines high susceptibility of murine A/J strain to iron-induced renal carcinogenesis, Cancer Sci. 113 (2021) 65–78.

[31] S. Toyokuni, Reactive oxygen species-induced molecular damage and its application in pathology, Pathol. Int. 49 (1999) 91–102.

[32] K. Yamashita, K. Kohashi, Y. Yamada, T. Ishii, Y. Nishida, H. Urakawa, I. Ito, M. Takahashi, T. Inoue, M. Ito, Y. Ohara, Y. Oda, S. Toyokuni, Osteogenic differentiation in dedifferentiated liposarcoma: a study of 36 cases in comparison to the cases without ossification, Histopathology 72 (5) (2018) 729–738.

[33] F. Ito, T. Nishiyama, L. Shi, M. Mori, T. Hirayama, H. Nagasawa, H. Yasui, S. Toyokuni, Contrasting intra- and extracellular distribution of catalytic ferrous iron in ovalbumin-induced peritonitis, Biochem. Biophys. Res. Commun. 476 (4) (2016) 600–606.

[34] H. Nagai, Y. Okazaki, S.H. Chew, N. Misawa, H. Yasui, S. Toyokuni, Deferasirox induces mesenchymal-epithelial transition in crocidolite-induced mesothelial carcinogenesis in rats, Cancer Prev. Res. 6 (2013) 1222–1230.

[35] L. Yue, Y. Luo, L. Jiang, Y. Sekido, S. Toyokuni, PCBP2 knockdown promotes ferroptosis in malignant mesothelioma, Pathol. Int. 72 (4) (2022) 242–251.

[36] R.A. Weinberg, The Biology of Cancer, 2 ed., Garland Science, Tailor & Francis Group, LLC, New York, 2014.

[37] G. Novara, G. Martignoni, W. Artibani, V. Ficarra, Grading systems in renal cell carcinoma, J. Urol. 177 (2) (2007) 430–436.

[38] M. Adebayo, S. Singh, A.P. Singh, S. Dasgupta, Mitochondrial fusion and fission: the fine-tune balance for cellular homeostasis, Faseb. J. 35 (6) (2021), e21620.

[39] M. Morita, S.P. Gravel, L. Hulea, O. Larsson, M. Pollak, J. St-Pierre, I. Topisirovic, mTOR coordinates protein synthesis, mitochondrial activity and proliferation, Cell Cycle 14 (4) (2015) 473–480.

[40] D.P. Panigrahi, P.P. Praharaj, C.S. Bhol, K.K. Mahapatra, S. Patra, B.P. Behera, S. R. Mishra, S.K. Bhutia, The emerging, multifaceted role of mitophagy in cancer and cancer therapeutics, Semin. Cancer Biol. 66 (2020) 45–58.

[41] S. Toyokuni, T. Tanaka, Y. Hattori, Y. Nishiyama, H. Ochi, H. Hiai, K. Uchida, T. Osawa, Quantitative immunohistochemical determination of 8-hydroxy-2’- deoxyguanosine by a monoclonal antibody N45.1: its application to ferric nitrilotriacetate-induced renal carcinogenesis model, Lab. Invest. 76 (1997) 365–374.

[42] L.J. Kuo, L.X. Yang, Gamma-H2AX - a novel biomarker for DNA double-strand breaks, in: Vivo, vol. 22, 2008, pp. 305–309, 3.

[43] J.R. Testa, M. Cheung, J. Pei, J.E. Below, Y. Tan, E. Sementino, N.J. Cox, A. U. Dogan, H.I. Pass, S. Trusa, M. Hesdorffer, M. Nasu, A. Powers, Z. Rivera, S. Comertpay, M. Tanji, G. Gaudino, H. Yang, M. Carbone, Germline BAP1 mutations predispose to malignant mesothelioma, Nat. Genet. 43 (10) (2011) 1022–1025.

[44] S. Pena-Llopis, ˜ S. Vega-Rubín-de-Celis, A. Liao, N. Leng, A. Pavía-Jim´enez, S. Wang, T. Yamasaki, L. Zhrebker, S. Sivanand, P. Spence, BAP1 loss defines a new class of renal cell carcinoma, Nat. Genet. 44 (7) (2012) 751.

[45] D.G. Albertson, Gene amplification in cancer, Trends Genet. 22 (8) (2006) 447–455.

[46] H. Kim, N.P. Nguyen, K. Turner, S. Wu, A.D. Gujar, J. Luebeck, J. Liu, V. Deshpande, U. Rajkumar, S. Namburi, S.B. Amin, E. Yi, F. Menghi, J.H. Schulte, A.G. Henssen, H.Y. Chang, C.R. Beck, P.S. Mischel, V. Bafna, R.G.W. Verhaak, Extrachromosomal DNA is associated with oncogene amplification and poor outcome across multiple cancers, Nat. Genet. 52 (9) (2020) 891–897.

[47] R.G.W. Verhaak, V. Bafna, P.S. Mischel, Extrachromosomal oncogene amplification in tumour pathogenesis and evolution, Nat. Rev. Cancer 19 (5) (2019) 283–288.

[48] R. Marcotte, A. Sayad, K.R. Brown, F. Sanchez-Garcia, J. Reimand, M. Haider, C. Virtanen, J.E. Bradner, G.D. Bader, G.B. Mills, D. Pe’er, J. Moffat, B.G. Neel, Functional genomic landscape of human breast cancer drivers, vulnerabilities, and resistance, Cell 164 (1–2) (2016) 293–309.

[49] Y. Chen, O.I. Olopade, MYC in breast tumor progression, Expert Rev. Anticancer Ther. 8 (10) (2008) 1689–1698.

[50] T.A. Grushko, J.J. Dignam, S. Das, A.M. Blackwood, C.M. Perou, K.K. Ridderstrale, K.N. Anderson, M.J. Wei, A.J. Adams, F.G. Hagos, L. Sveen, H.T. Lynch, B. L. Weber, O.I. Olopade, MYC is amplified in BRCA1-associated breast cancers, Clin. Cancer Res. 10 (2) (2004) 499–507.

[51] Q. Wang, H. Zhang, K. Kajino, M.I. Greene, BRCA1 binds c-Myc and inhibits its transcriptional and transforming activity in cells, Oncogene 17 (15) (1998) 1939–1948.

[52] R.D. Kennedy, J.J. Gorski, J.E. Quinn, G.E. Stewart, C.R. James, S. Moore, K. Mulligan, E.D. Emberley, T.F. Lioe, P.J. Morrison, P.B. Mullan, G. Reid, P. G. Johnston, P.H. Watson, D.P. Harkin, BRCA1 and c-Myc associate to transcriptionally repress psoriasin, a DNA damage-inducible gene, Cancer Res. 65 (22) (2005) 10265–10272.

[53] R.L. Idzerda, H. Huebers, C.A. Finch, G.S. McKnight, Rat transferrin gene expression: tissue-specific regulation by iron deficiency, Proc. Natl. Acad. Sci. U.S. A. 83 (11) (1986) 3723–3727.

[54] A.G. Casanova, L. Vicente-Vicente, M.T. Hernandez-Sanchez, M. Prieto, M. I. Rihuete, L.M. Ramis, E. Del Barco, J.J. Cruz, A. Ortiz, I. Cruz-Gonzalez, C. Martinez-Salgado, M. Pescador, F.J. Lopez-Hernandez, A.I. Morales, Urinary transferrin pre-emptively identifies the risk of renal damage posed by subclinical tubular alterations, Biomed. Pharmacother. 121 (2020) 109684.

[55] B.R. Stockwell, J.P. Friedmann Angeli, H. Bayir, A.I. Bush, M. Conrad, S.J. Dixon, S. Fulda, S. Gascon, S.K. Hatzios, V.E. Kagan, K. Noel, X. Jiang, A. Linkermann, M. E. Murphy, M. Overholtzer, A. Oyagi, G.C. Pagnussat, J. Park, Q. Ran, C. S. Rosenfeld, K. Salnikow, D. Tang, F.M. Torti, S.V. Torti, S. Toyokuni, K. A. Woerpel, D.D. Zhang, Ferroptosis: a regulated cell death nexus linking metabolism, Redox Biol. Dise. Cell. 171 (2) (2017) 273–285.

[56] S. Okada, S. Hamazaki, Y. Ebina, J.-L. Li, O. Midorikawa, Nephrotoxicity and its prevention by vitamin E on ferric nitrilotriacetate-promoted lipid peroxidation, Biochim. Biophys. Acta 922 (1987) 28–33.

[57] S. Hamazaki, S. Okada, Y. Ebina, J.L. Li, O. Midorikawa, Effect of dietary vitamin E on ferric nitrilotriacetate-induced nephrotoxicity in rats, Toxicol. Appl. Pharmacol. 92 (1988) 500–506.

[58] M. Whitnall, Y. Suryo Rahmanto, M.L. Huang, F. Saletta, H.C. Lok, L. Gutierrez, F. J. Lazaro, A.J. Fleming, T.G. St Pierre, M.R. Mikhael, P. Ponka, D.R. Richardson, Identification of nonferritin mitochondrial iron deposits in a mouse model of Friedreich ataxia, Proc. Natl. Acad. Sci. U.S.A. 109 (50) (2012) 20590–20595.

[59] J.J. Braymer, R. Lill, Iron-sulfur cluster biogenesis and trafficking in mitochondria, J. Biol. Chem. 292 (31) (2017) 12754–12763.

[60] B.T. Paul, D.H. Manz, F.M. Torti, S.V. Torti, Mitochondria and Iron: current questions, Expet Rev. Hematol. 10 (1) (2017) 65–79.

[61] D. Hanahan, R.A. Weinberg, Hallmarks of cancer: the next generation, Cell 144 (5) (2011) 646–674.

[62] I. Yanatori, D.R. Richardson, S. Toyokuni, F. Kishi, The iron chaperone poly(rC)- binding protein 2 forms a metabolon with the heme oxygenase 1/cytochrome P450 reductase complex for heme catabolism and iron transfer, J. Biol. Chem. 292 (32) (2017) 13205–13229.

[63] I. Yanatori, D.R. Richardson, S. Toyokuni, F. Kishi, The new role of poly (rC)- binding proteins as iron transport chaperones: proteins that could couple with inter-organelle interactions to safely traffic iron, Biochim. Biophys. Acta Gen. Subj. 1864 (11) (2020) 129685.

[64] D.R. Fraser, E. Kodicek, Unique biosynthesis by kidney of a biological active vitamin D metabolite, Nature 228 (5273) (1970) 764–766.

[65] J.G. Ghazarian, C.R. Jefcoate, J.C. Knutson, W.H. Orme-Johnson, H.F. DeLuca, Mitochondrial cytochrome p450. A component of chick kidney 25- hydrocholecalciferol-1alpha-hydroxylase, J. Biol. Chem. 249 (10) (1974) 3026–3033.

[66] W.B. Grant, A review of the evidence supporting the vitamin D-cancer prevention hypothesis in 2017, Anticancer Res. 38 (2) (2018) 1121–1136.

[67] S.A. Krum, E. la Rosa Dalugdugan, G.A. Miranda-Carboni, T.F. Lane, BRCA1 forms a functional complex with gamma-H2AX as a late response to genotoxic stress, J. Nucleic Acids 2010 (2010) 801594.

[68] K.I. Savage, K.B. Matchett, E.M. Barros, K.M. Cooper, G.W. Irwin, J.J. Gorski, K. S. Orr, J. Vohhodina, J.N. Kavanagh, A.F. Madden, A. Powell, L. Manti, S. S. McDade, B.H. Park, K.M. Prise, S.A. McIntosh, M. Salto-Tellez, D.J. Richard, C. T. Elliott, D.P. Harkin, BRCA1 deficiency exacerbates estrogen-induced DNA damage and genomic instability, Cancer Res. 74 (10) (2014) 2773–2784.

[69] R.C. West, G.J. Bouma, Q.A. Winger, Shifting perspectives from "oncogenic" to oncofetal proteins; how these factors drive placental development, Reprod. Biol. Endocrinol. 16 (1) (2018) 101.

[70] S.Y. Lin, K. Li, G.S. Stewart, S.J. Elledge, Human Claspin works with BRCA1 to both positively and negatively regulate cell proliferation, Proc. Natl. Acad. Sci. U.S.A. 101 (17) (2004) 6484–6489.

[71] N. Izawa, W. Wu, K. Sato, H. Nishikawa, A. Kato, N. Boku, F. Itoh, T. Ohta, HERC2 Interacts with Claspin and regulates DNA origin firing and replication fork progression, Cancer Res. 71 (17) (2011) 5621–5625.

[72] R.I. Yarden, S. Pardo-Reoyo, M. Sgagias, K.H. Cowan, L.C. Brody, BRCA1 regulates the G2/M checkpoint by activating Chk1 kinase upon DNA damage, Nat. Genet. 30 (3) (2002) 285–289.

[73] D. Cortez, Y. Wang, J. Qin, S.J. Elledge, Requirement of ATM-dependent phosphorylation of brca1 in the DNA damage response to double-strand breaks, Science 286 (5442) (1999) 1162–1166.

[74] N. Foray, D. Marot, A. Gabriel, V. Randrianarison, A.M. Carr, M. Perricaudet, A. Ashworth, P. Jeggo, A subset of ATM- and ATR-dependent phosphorylation events requires the BRCA1 protein, EMBO J. 22 (11) (2003) 2860–2871.

[75] R.S. Tibbetts, D. Cortez, K.M. Brumbaugh, R. Scully, D. Livingston, S.J. Elledge, R. T. Abraham, Functional interactions between BRCA1 and the checkpoint kinase ATR during genotoxic stress, Genes Dev. 14 (23) (2000) 2989–3002.

[76] Y. Ohara, S.H. Chew, T. Shibata, Y. Okazaki, K. Yamashita, S. Toyokuni, Phlebotomy as a preventive measure for crocidolite-induced mesothelioma in male rats, Cancer Sci. 109 (2) (2018) 330–339.

参考文献をもっと見る