Skip to main content

Environmental impact on carcinogenesis under BRCA1 haploinsufficiency

Abstract

Cancer is the primary cause of human mortality in Japan since 1981. Although numerous novel therapies have been developed and applied in clinics, the number of deaths from cancer is still increasing worldwide. It is time to consider the strategy of cancer prevention more seriously. Here we propose a hypothesis that cancer can be side effects of long time-use of iron and oxygen and that carcinogenesis is an evolution-like cellular events to obtain “iron addiction with ferroptosis-resistance” where genes and environment interact each other. Among the recognized genetic risk factors for carcinogenesis, we here focus on BRCA1 tumor suppressor gene and how environmental factors, including daily life exposure and diets, may impact toward carcinogenesis under BRCA1 haploinsufficiency. Although mice models of BRCA1 mutants have not been successful for decades in generating phenotype mimicking the human counterparts, a rat model of BRCA1 mutant was recently established that reasonably mimics the human phenotype. Two distinct categories of oxidative stress, one by radiation and one by iron-catalyzed Fenton reaction, promoted carcinogenesis in Brca1 rat mutants. Furthermore, mitochondrial damage followed by alteration of iron metabolism finally resulted in ferroptosis-resistance of target cells in carcinogenesis. These suggest a possibility that cancer prevention by active pharmacological intervention may be possible for BRCA1 mutants to increase the quality of their life rather than preventive mastectomy and/or oophorectomy.

Introduction

Cancer is the leading cause of human mortality in Japan since 1981 (https://ganjoho.jp/public/qa_links/report/statistics/2022_en.html). Although numerous novel therapies, such as immune checkpoint inhibitors [1] and chimeric antigen receptor T-cell therapy [2], have been developed and applied in clinics recently, the number of deaths from cancer is still increasing worldwide (https://www.who.int/news-room/fact-sheets/detail/cancer). It is time to consider the strategies for cancer prevention more seriously and comprehensively to decrease the burden to the society.

We have been proposing a hypothesis that cancer can be the side effects of long-time use of iron and oxygen [3] if we can eliminate the established risks, including physical, chemical and biological carcinogens (https://www.who.int/news-room/fact-sheets/detail/cancer) and that carcinogenesis is generally a process to obtain “iron addiction with ferroptosis-resistance” [4]. The proof of concept for this hypothesis is that iron is indispensable for cell proliferation to replicate DNA [5] and that Fe(II) is a catalyst for the Fenton reaction, which generates the most damaging and mutagenic chemical species, hydroxyl radical [6]. This is further based on our own observation and observation by other investigators that 1) excess iron in various human pathology is associated with higher risk for carcinogenesis [7,8,9]; 2) iron reduction by phlebotomy decreases the cancer risk and mortality in a human intervention study [10]; 3) repeated iron-catalyzed Fenton reaction causes aggressive cancer that is similar to human counterparts not only in macroscopic/microscopic morphology but also in genetic alterations [11, 12]. These animal models include ferric nitrilotriacetate (Fe-NTA)-induced renal carcinogenesis [12,13,14,15] and asbestos-induced mesothelial carcinogenesis in rats [16,17,18,19]; 4) especially in the latter case, iron removal by iron chelating agent [20] or phlebotomy [21] can prevent mesothelial carcinogenesis to some extent. More detailed review on these topics are found elsewhere [9, 12, 22]. At first, we here describe the recent advances in iron metabolism in mammals, including the concept of ferroptosis.

Recent advances in iron metabolism

Iron is the most abundant heavy metal in our body and is indispensable for all the lives on earth [7, 23, 24]. Iron basically works in two ways in higher mammals: 1) persistent electron transfer via redox cycling and 2) temporary oxygen storage as heme in hemoglobin, myoglobin, neuroglobin and cytoglobin. Indeed, ~ 60% of iron is in hemoglobin in humans. Because iron is thus important, our body is deficient of any active mechanism to discharge iron to outside our body [25].

Serum iron-transporting protein, transferrin, has been recognized since 1946 [26] and transferrin receptor 1 was identified in 1981 [26]. Iron storage protein ferritin was cloned in 1986 [27]. However, it took some extra time for membrane iron transporters and iron chaperones to be established [28, 29]. Of note, Fe(III) is insoluble at neutral pH and used for extracellular transport and intracellular storage (Fig. 1). In contrast, Fe(II) is soluble and used for transport across the membrane and intracellular transport. Labile iron is a concept indicating cytosolic mobile free iron [30], but some ambiguity still remains in that labile iron includes catalytic Fe(II), chaperoned Fe(II) by poly(rC) binding protein 1/2 (PCBP1/2) [31, 32] and dinitrosyl-diglutathionyl iron complex (DNDGIC) [33]. Figure 2 shows the current summary of iron metabolism.

Fig. 1
figure 1

Difference in the biological significance of Fe(II) and Fe(III). DNDGIC, dinitrosyl-diglutathionyl-iron complex

Fig. 2
figure 2

Current understanding of iron metabolism. Recently, many novel concepts have been established regarding iron metabolism, including ferritinophagy to take out iron from ferritin, cytosolic iron chaperones, PCBP1/2 and Fe(III)-loaded ferritin release via CD63-regulated exosomes. CPN, ceruloplasmin; Dcytb, duodenal cytochrome B; DMT1, divalent metal transporter-1 (SLC11A2); FPN, ferroportin (SLC40A1); TF, transferrin; STEAP3, six-transmembrane epithelial antigen of the prostate; TfR1, transferrin receptor-1; PCBP, poly(rC) binding protein; IRE-IRP, iron-responsive element-iron regulatory protein; brown circle as Fe(II); blue circle as Fe(III); green letter, reductase; pink letter, oxidase

A recent new finding in iron metabolism is that our cells use exosomes for the monopoly of iron inside ourselves [34]. The importance of iron for survival is the same for other microorganisms, such as bacteria, fungi and parasites. Those infectious agents try to steal iron from our cells. They use many different molecules, including siderophores [35]. Interestingly, one of the siderophores of a bacterium, desferrioxiamine, is used as an iron-chelating agent for medical use [36]. We recently found that a characteristic membrane surface molecule on exosome, CD63, is under the regulation of iron-responsive element/iron-regulatory protein (IRE/IRP) system [34]. This posttranscriptional regulatory system is specific for iron metabolism and IRE sequence is observed either in the 5’ or 3’ portion of mRNA of iron metabolism-associated genes. This is a system for iron deficiency (Fig. 2), considering the era of hunger [37]. Transferrin receptor 1 (Tfr1) mRNA has 5 IREs in the 3’ portions, thus increasing the lifetime of this message to increase the amounts of the Tfr1 protein. Conversely, translation of iron storage protein Fth1/Ftl is blocked for translation when the cell is iron-deficient. In the case of CD63, IRE sequence is present at the 5’ portion. If the cells harbor ample amounts of iron, this will be deblocked and exosomes with iron-loaded ferritin is generated through nuclear receptor activator 4 (NCOA4) and secreted toward the other cells of the same individual. Indeed, this is a safe strategy to transfer surplus iron to neighbor iron-deficient cells. Here we would like to stress that this IRE sequence in CD63 is present only in higher primates and is not present in mice or rats, which are used for experiments. However, this system is abused in asbestos-induced mesothelial carcinogenesis [22, 38, 39].

Ferroptosis

There are only two types of cell death classified by light microscopy, necrosis and apoptosis. However, starting from the 2000’s, many cell death modes were proposed, defined by the specific signaling pathways. These include ferroptosis (Fig. 3), catalytic Fe(II)-regulated necrosis accompanied by lipid peroxidation [40, 41]. Ferroptosis just celebrated its 10th birthday in 2022, and this cell mode became popular evidenced by the exponentially increasing number of papers studying ferroptosis [42].

Fig. 3
figure 3

Current understanding of ferroptosis, catalytic Fe(II)-dependent regulated necrosis associated with lipid peroxidation. Ferroptosis is three dimensionally regulated by Fe, S and O. Transition to high Fe/S ratio by certain stimulus (Ex. excess iron and erastin [inhibitor of cystine/glutamate antiporter]) to cells initiate uncontrollable lipid peroxidation, which is cellular catalytic Fe(II) dependent and designated as ferroptosis. Ferroptotic cells reveal the morphology of necrosis. ACSL4, acyl-CoA synthatase long-chain 4; GPX4, glutathione peroxidase-4; PUFAs, polyunsaturated fatty acids

We have been working on iron-induced carcinogenesis for decades. Among them, repeated intraperitoneal injections of ferric nitrilotriacetate (Fe-NTA) induces renal cell carcinoma (RCC) in a high incidence (60 ~ 90%) in male rats [9, 12]. In this model, renal tubular necrosis is observed as early as 30 min in the proximal tubular cells with various lipid peroxidation products [43,44,45,46], which are the observation of our own in the 1980’s and 1990’s. When we first recognized the word ferroptosis in 2014, we immediately accepted it, based on our experience of this oxidative renal tubular damage. In the Fe-NTA-induced renal carcinogenesis model, renal tubular cells obtain ferroptosis-resistance in a few weeks after continued iron-catalyzed oxidative stress [12, 47, 48]. We have been proposing a hypothesis that carcinogenesis is a process to acquire ferroptosis-resistance under iron addiction via somatic mutation(s) [4]. Iron is an essential cofactor for ribonucleotide reductase for DNA synthesis and replication [49, 50], which is indispensable for proliferating cancer cells.

Accordingly, cancer cells harbor higher amounts of catalytic Fe(II) in the cytosol in comparison to the non-tumorous cells [51, 52]. This high amounts of catalytic Fe(II) is useful for DNA replication but also causes persistent oxidative stress to the cancer cells [53]. Thus, cancer cells are prepared to counteract this oxidative stress, for example, via the activation of Nrf2 transcription factor, a master regulator of antioxidative genes [54, 55]. Ferroptosis may be interpreted as relative predominance of iron over sulfur (sulfhydryls) by stimuli, which is modulated by the amounts of polyunsaturated fatty acids (PUFAs), mainly as phospholipids, in the cellular membrane (Fig. 3). This is indeed the Achilles’ heels of cancer cells and numerous ferroptosis inducers are currently under investigation for cancer therapy [3, 5, 56].

Alternatively, we recently found physiological ferroptosis. We selected a mouse monoclonal antibody for 4-hydroxy-2-nonenal (HNE)-modified proteins, HNEJ-1 clone, to detect ferroptotic cells in formalin-fixed paraffin-embedded specimens [57, 58]. Our present conclusion is that ferroptotic event occurs in nucleated red blood cells at E13.5 and aging cells of various organs in rats [59]. Thus, it is not strange to find ferroptosis in neuronal cells in various neurogenerative diseases [60,61,62]. Here researchers are trying to stop ferroptosis in the dying neuronal cells by developing agents to prevent ferroptosis. In summary, ferroptosis is now an optimal target for the development of new drugs both for induction and prevention.

BRCA1

Current understanding is that cancer is a disease of the genome [63]. Thus far, we suggested that iron and oxygen can be the major mutagens in the long human lifetime of more than 80 years [3, 64]. Other than iron and oxygen, there are a plethora of mutagenic agents exposed to humans via skin, respiratory tract or gastrointestinal tract, which are both natural and industrial (https://monographs.iarc.who.int/agents-classified-by-the-iarc/). On the other hand, genetic susceptibility of each individual is as important as mutagens because there would be no carcinogenesis if the prevention and repair processes are perfect. Various familial cancer syndromes have been recognized from long time ago [63]. Since 1990’s, tumor suppressor genes were identified and cloned one by one [65]. These were the genes for the repair of various damage to genomic DNA or the genes to cause cell death with defined levels of biological/chemical/physical stimulus or damage. One of the most socially recognized tumor suppressor genes is BRCA1 due to the famous Hollywood actress, Angelina Jolie, known as the Angelina effect [66, 67].

Here according to a recent report on the Japanese population, the target organs for carcinogenesis of BRCA1 mutants include female breast (odds ratio [OR], 16.1; p = 3.50 × 10–11) and ovary (OR, 75.6; p = 2.26 × 10–22) [68], which are critical for reproduction as the nutrient source of for the next generation and the reserve of oocytes, respectively. The present guideline still recommends prophylactic mastectomy [69] and oophorectomy [70] when necessary, which has been sensational to the general public. A higher risk for biliary tract cancer (OR, 17.4; p = 2.96 × 10–7), pancreatic cancer (OR, 12.6; p = 4.67 × 10–5) and gastric cancer (OR, 5.2; p = 3.40 × 10–6) is also noted recently for BRCA1 mutants [68]. Considering the characteristics of target organs in BRCA1-associated carcinogenesis, we hypothesized that iron-associated oxidative stress may be in common as a promotional factor, especially for breast and ovary. This is based on the fact that both organs are deeply associated with iron metabolism including lactoferrin secretion in milk [71, 72] and ovulation. Ovarian endometriosis is closely associated with ovarian carcinoma through iron-mediated oxidative stress [73,74,75,76]. If so, some other preventive strategies may be possible.

Species difference in animal experiment

BRCA1 tumor suppressor gene was cloned in 1994 by Miki et al. [77]. Thereafter, hundreds of trials were performed to generate a feasible murine model of human BRCA1 mutants. However, this was not successful in that heterozygous knockout of BRCA1 alone showed no phenotype in carcinogenesis whereas homozygous knockout was embryonic lethal [78]. Many conditional knockout mice model was produced, but the results were negative. If the heterozygous knockout mice were crossed with TP53( ±) mice, the mice showed susceptibility to basal-like breast cancer [79].

However, it was surprising that rat Brca1 mutant model (L63X/ +) shows the phenotype. This model was developed by Imaoka and Mashimo et al. in 2022 in Japan [80]. We believe that this is a species difference and that Rattus norvegicus is significantly closer to Homo sapiens in comparison to Mus musculus. We thus far observed similar phenomena in Fe-NTA-induced renal carcinogenesis. Whereas renal carcinogenesis is observed in mice and rats, phenotypes are quite different (Table 1), which is much milder in mice in comparison to rats [81].

Table 1 Species differences in ferric nitrilotriacetate (Fe-NTA)-induced renal cell carcinoma in rodents

BRCA1 and ferroptosis-resistance

We have recently applied Fe-NTA renal carcinogenesis model to male Brca1(L63X/ +) rats to evaluate whether iron-catalyzed oxidative stress [12] is important for Brca1 mutant carcinogenesis [83]. The incidence of renal carcinogenesis was not changed between the Brca1 mutant and the wild-type. However, the carcinogenesis was significantly promoted in the Brca1 mutants by 3 months on average in comparison to the wild-type, which is a marked difference considering the average life time rats of ~ 3 y. This result indicates that iron-catalyzed oxidative stress is a promoting factor of carcinogenesis for Brca1 mutants.

Furthermore, we found that renal cell carcinomas (RCCs) in Brca1 mutants show more genomic alterations, including c-Myc amplification [83], which is indeed frequently observed in the breast carcinoma of human BRCA1 mutants [87] and is a risk for poor prognosis [88, 89]. Here c-Myc amplification was often extrachromosomal. These results suggest that iron removal or avoidance of oxidative stress in the target organs could be an effective measure to prevent carcinogenesis in BRCA1 mutants.

We then undertook to understand the molecular mechanism why iron-catalyzed oxidative stress promotes renal carcinogenesis. We have performed expression microarray analysis in the subacute phase of 3 weeks during the renal carcinogenesis and found that higher mitochondrial damage is a key phenomenon [83]. Electron microscopical analysis revealed that even the untreated control kidney showed smaller and deformed mitochondria in the renal tubular cells of Brca1 mutants. Since mitochondria play a central role in iron metabolism producing heme, it is plausible that mitochondrial damage alters iron metabolism in the entire cell, which produced a niche for carcinogenesis under mutagenic environment with Fe(III) abundance but with less catalytic Fe(II) at the subacute phase in the Brca1 mutants in comparison to the wild-type. This is the mechanism how iron addiction with ferroptosis-resistance was generated (Fig. 4). We recently obtained similar results on chrysotile-induced malignant mesothelioma by the use of male Brca1(L63X/ +) rats [90]. However, we still need to know the role of Brca1 haploinsufficiency in this mitochondrial damage and the demonstration in human BRCA1 mutant samples would be necessary.

Fig. 4
figure 4

Role of BRCA1 haploinsufficiency in carcinogenesis. BRCA1 haploinsufficiency causes iron-rich mutagenic environment under persistent oxidative stress mediate by iron to promote carcinogenesis through ferroptosis-resistance and allows c-Myc amplification

Conclusion

Cancer is basically a disease of the genome, where genome and environment persistently interact each other. We believe that even the long-use of iron and oxygen eventually causes various mutations, which may explain why aging stands as one of the highest risks for cancer. Some of the cancer susceptibility can be explained by the inactivation of tumor suppressor genes. Here this review article focused on how we undertook to find promoting factors in BRCA1 mutants with a recently established rat Brca1(L63X/ +) model. During iron-induced renal carcinogenesis, Brca1 haploinsufficiency allowed more genomic alterations, including amplification of c-Myc. Therefore, environmental factors, such as the control of iron and oxidative stress, may work as a strategy to prevent or delay carcinogenesis in BRCA1 mutants.

Availability of data and materials

Not applicable.

Abbreviations

DNDGIC:

Dinitrosyl-diglutathionyl iron complex

Fe-NTA:

Ferric nitrilotriacetate

HNE:

4-Hydroxy-2-nonenal

IRE/IRP:

Iron-responsive element/iron-regulatory protein

NCOA4:

Nuclear receptor activator 4

OR:

Odds radio

PCBP1/2:

Poly(rC) binding protein 1/2

PUFAs:

Polyunsaturated fatty acids

RCC:

Renal cell carcinoma

References

  1. Sharma P, Allison JP. Dissecting the mechanisms of immune checkpoint therapy. Nat Rev Immunol. 2020;20(2):75–6.

    Article  CAS  Google Scholar 

  2. Neelapu SS, Tummala S, Kebriaei P, Wierda W, Gutierrez C, Locke FL, et al. Chimeric antigen receptor T-cell therapy - assessment and management of toxicities. Nat Rev Clin Oncol. 2018;15(1):47–62.

    Article  CAS  Google Scholar 

  3. Toyokuni S, Kong Y, Cheng Z, Sato K, Hayashi S, Ito F, et al. Carcinogenesis as Side Effects of Iron and Oxygen Utilization: From the Unveiled Truth toward Ultimate Bioengineering. Cancers (Basel). 2020;12(11):3320.

    Article  CAS  Google Scholar 

  4. Toyokuni S, Ito F, Yamashita K, Okazaki Y, Akatsuka S. Iron and thiol redox signaling in cancer: An exquisite balance to escape ferroptosis. Free Radic Biol Med. 2017;108:610–26.

    Article  CAS  Google Scholar 

  5. Toyokuni S, Yanatori I, Kong Y, Zheng H, Motooka Y, Jiang L. Ferroptosis at the crossroads of infection, aging and cancer. Cancer Sci. 2020;111:2665–71.

    Article  CAS  Google Scholar 

  6. Koppenol WH, Hider RH. Iron and redox cycling Do’s and don’ts. Free Radic Biol Med. 2019;133:3–10.

    Article  CAS  Google Scholar 

  7. Toyokuni S. Iron-induced carcinogenesis: the role of redox regulation. Free Radic Biol Med. 1996;20:553–66.

    Article  CAS  Google Scholar 

  8. Toyokuni S. Iron and thiols as two major players in carcinogenesis: friends or foes? Front Pharmacol. 2014;5:200.

    Article  Google Scholar 

  9. Toyokuni S. 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. 2016;66:245–59.

    Article  CAS  Google Scholar 

  10. Zacharski L, Chow B, Howes P, Shamayeva G, Baron J, Dalman R, et al. Decreased cancer risk after iron reduction in patients with peripheral arterial disease: Results from a randomized trial. J Natl Cancer Inst. 2008;100:996–1002.

    Article  CAS  Google Scholar 

  11. Akatsuka S, Yamashita Y, Ohara H, Liu YT, Izumiya M, Abe K, et al. Fenton reaction induced cancer in wild type rats recapitulates genomic alterations observed in human cancer. PLoS ONE. 2012;7(8): e43403.

    Article  CAS  Google Scholar 

  12. Toyokuni S, Kong Y, Zheng H, Maeda Y, Motooka Y, Akatsuka S. Iron as spirit of life to share under monopoly. J Clin Biochem Nutr. 2022;71(2):78–88.

    Article  CAS  Google Scholar 

  13. Ebina Y, Okada S, Hamazaki S, Ogino F, Li JL, Midorikawa O. Nephrotoxicity and renal cell carcinoma after use of iron- and aluminum- nitrilotriacetate complexes in rats. J Natl Cancer Inst. 1986;76:107–13.

    CAS  Google Scholar 

  14. Li JL, Okada S, Hamazaki S, Ebina Y, Midorikawa O. Subacute nephrotoxicity and induction of renal cell carcinoma in mice treated with ferric nitrilotriacetate. Cancer Res. 1987;47:1867–9.

    CAS  Google Scholar 

  15. Nishiyama Y, Suwa H, Okamoto K, Fukumoto M, Hiai H, Toyokuni S. 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. 1995;86:1150–8.

    Article  CAS  Google Scholar 

  16. Toyokuni S. Mechanisms of asbestos-induced carcinogenesis. Nagoya J Med Sci. 2009;71(1–2):1–10.

    CAS  Google Scholar 

  17. Jiang L, Akatsuka S, Nagai H, Chew SH, Ohara H, Okazaki Y, et al. Iron overload signature in chrysotile-induced malignant mesothelioma. J Pathol. 2012;228:366–77.

    Article  CAS  Google Scholar 

  18. Toyokuni S. Iron addiction with ferroptosis-resistance in asbestos-induced mesothelial carcinogenesis: Toward the era of mesothelioma prevention. Free Radic Biol Med. 2019;133:206–15.

    Article  CAS  Google Scholar 

  19. Toyokuni S, Ito F, Motooka Y. Role of ferroptosis in nanofiber-induced carcinogenesis. Metallomics Res. 2021;1(1):14–21.

    Google Scholar 

  20. Nagai H, Okazaki Y, Chew SH, Misawa N, Yasui H, Toyokuni S. Deferasirox induces mesenchymal-epithelial transition in crocidolite-induced mesothelial carcinogenesis in rats. Cancer Prev Res (Phila). 2013;6:1222–30.

    Article  CAS  Google Scholar 

  21. Ohara Y, Chew SH, Shibata T, Okazaki Y, Yamashita K, Toyokuni S. Phlebotomy as a preventive measure for crocidolite-induced mesothelioma in male rats. Cancer Sci. 2018;109(2):330–9.

    Article  CAS  Google Scholar 

  22. Toyokuni S, Kong Y, Zheng H, Mi D, Katabuchi M, Motooka Y, et al. Double-edged Sword Role of Iron-loaded Ferritin in Extracellular Vesicles. J Cancer Prev. 2021;26(4):244–9.

    Article  Google Scholar 

  23. Torti SV, Torti FM. Iron and cancer: more ore to be mined. Nat Rev Cancer. 2013;13(5):342–55.

    Article  CAS  Google Scholar 

  24. Drakesmith H, Nemeth E, Ganz T. Ironing out Ferroportin. Cell Metab. 2015;22(5):777–87.

    Article  CAS  Google Scholar 

  25. Toyokuni S. Role of iron in carcinogenesis: Cancer as a ferrotoxic disease. Cancer Sci. 2009;100(1):9–16.

    Article  CAS  Google Scholar 

  26. Sutherland R, Delia D, Schneider C, Newman R, Kemshead J, Greaves M. Ubiquitous cell-surface glycoprotein on tumor cells is proliferation-associated receptor for transferrin. Proc Natl Acad Sci U S A. 1981;78(7):4515–9.

    Article  CAS  Google Scholar 

  27. Hentze MW, Keim S, Papadopoulos P, O’Brien S, Modi W, Drysdale J, et al. Cloning, characterization, expression, and chromosomal localization of a human ferritin heavy-chain gene. Proc Natl Acad Sci U S A. 1986;83(19):7226–30.

    Article  CAS  Google Scholar 

  28. Gunshin H, Mackenzie B, Berger U, Gunshin Y, Romero M, Boron W, et al. Cloning and characterization of a mammalian proton-coupled metal-ion transporter. Nature. 1997;388(6641):482–8.

    Article  CAS  Google Scholar 

  29. Donovan A, Brownlie A, Zhou Y, Shepard J, Pratt S, Moynihan J, et al. Positional cloning of zebrafish ferroportin1 identifies a conserved vertebrate iron exporter. Nature. 2000;403(6771):776–81.

    Article  CAS  Google Scholar 

  30. Gutteridge J, Rowley D, Halliwell B. Superoxide-dependent formation of hydroxyl radicals in the presence of iron salts Detection of “free” iron in biological systems by using bleomycin-dependent degradation of DNA. Biochem J. 1981;199(1):263–5.

    Article  CAS  Google Scholar 

  31. Yanatori I, Richardson DR, Toyokuni S, Kishi F. 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. 2017;292(32):13205–29.

    Article  CAS  Google Scholar 

  32. Yanatori I, Richardson DR, Toyokuni S, Kishi F. 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. 2020;1864(11): 129685.

    Article  CAS  Google Scholar 

  33. Richardson DR, Lok HC. The nitric oxide-iron interplay in mammalian cells: transport and storage of dinitrosyl iron complexes. Biochim Biophys Acta. 2008;1780(4):638–51.

    Article  CAS  Google Scholar 

  34. Yanatori I, Richardson DR, Dhekne HS, Toyokuni S, Kishi F. CD63 is regulated by iron via the IRE-IRP system and is important for ferritin secretion by extracellular vesicles. Blood. 2021;138(16):1490–503.

    Article  CAS  Google Scholar 

  35. Winkelmann G. Microbial siderophore-mediated transport. Biochem Soc Trans. 2002;30(4):691–6.

    Article  CAS  Google Scholar 

  36. Codd R, Richardson-Sanchez T, Telfer TJ, Gotsbacher MP. Advances in the Chemical Biology of Desferrioxamine B. ACS Chem Biol. 2018;13(1):11–25.

    Article  CAS  Google Scholar 

  37. Muckenthaler MU, Galy B, Hentze MW. Systemic iron homeostasis and the iron-responsive element/iron-regulatory protein (IRE/IRP) regulatory network. Annu Rev Nutr. 2008;28:197–213.

    Article  CAS  Google Scholar 

  38. Ito F, Yanatori I, Maeda Y, Nimura K, Ito S, Hirayama T, et al. Asbestos conceives Fe(II)-dependent mutagenic stromal milieu through ceaseless macrophage ferroptosis and beta-catenin induction in mesothelium. Redox Biol. 2020;36: 101616.

    Article  CAS  Google Scholar 

  39. Ito F, Kato K, Yanatori I, Murohara T, Toyokuni S. Ferroptosis-dependent extracellular vesicles from macrophage contribute to asbestos-induced mesothelial carcinogenesis through loading ferritin. Redox Biol. 2021;47: 102174.

    Article  CAS  Google Scholar 

  40. Dixon SJ, Lemberg KM, Lamprecht MR, Skouta R, Zaitsev EM, Gleason CE, et al. Ferroptosis: an iron-dependent form of nonapoptotic cell death. Cell. 2012;149(5):1060–72.

    Article  CAS  Google Scholar 

  41. Stockwell BR, Friedmann Angeli JP, Bayir H, Bush AI, Conrad M, Dixon SJ, et al. Ferroptosis: A Regulated Cell Death Nexus Linking Metabolism, Redox Biology, and Disease. Cell. 2017;171(2):273–85.

    Article  CAS  Google Scholar 

  42. Stockwell BR. Ferroptosis turns 10: Emerging mechanisms, physiological functions, and therapeutic applications. Cell. 2022;185(14):2401–21.

    Article  CAS  Google Scholar 

  43. Hamazaki S, Okada S, Ebina Y, Midorikawa O. Acute renal failure and glucosuria induced by ferric nitrilotriacetate in rats. Toxicol Appl Pharmacol. 1985;77:267–74.

    Article  CAS  Google Scholar 

  44. Toyokuni S, Uchida K, Okamoto K, Hattori-Nakakuki Y, Hiai H, Stadtman ER. 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 USA. 1994;91:2616–20.

    Article  CAS  Google Scholar 

  45. Toyokuni S, Luo XP, Tanaka T, Uchida K, Hiai H, Lehotay DC. 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. 1997;22:1019–27.

    Article  CAS  Google Scholar 

  46. Kawai Y, Furuhata A, Toyokuni S, Aratani Y, Uchida K. Formation of acrolein-derived 2’-deoxyadenosine adduct in an iron-induced carcinogenesis model. J Biol Chem. 2003;278(50):50346–54.

    Article  CAS  Google Scholar 

  47. Tanaka T, Kondo S, Iwasa Y, Hiai H, Toyokuni S. Expression of stress-response and cell proliferation genes in renal cell carcinoma induced by oxidative stress. Am J Pathol. 2000;156(6):2149–57.

    Article  CAS  Google Scholar 

  48. Hiroyasu M, Ozeki M, Kohda H, Echizenya M, Tanaka T, Hiai H, et al. Specific allelic loss of p16 (INK4A) tumor suppressor gene after weeks of iron-mediated oxidative damage during rat renal carcinogenesis. Am J Pathol. 2002;160(2):419–24.

    Article  CAS  Google Scholar 

  49. Bollinger JM Jr, Edmondson DE, Huynh BH, Filley J, Norton JR, Stubbe J. Mechanism of assembly of the tyrosyl radical-dinuclear iron cluster cofactor of ribonucleotide reductase. Science. 1991;253(5017):292–8.

    Article  CAS  Google Scholar 

  50. Cotruvo JA, Stubbe J. Class I Ribonucleotide Reductases: Metallocofactor Assembly and Repair In Vitro and In Vivo. Ann Rev Biochem. 2011;80:733–67.

    Article  CAS  Google Scholar 

  51. Ito F, Nishiyama T, Shi L, Mori M, Hirayama T, Nagasawa H, et al. Contrasting intra- and extracellular distribution of catalytic ferrous iron in ovalbumin-induced peritonitis. Biochem Biophys Res Commun. 2016;476(4):600–6.

    Article  CAS  Google Scholar 

  52. Schoenfeld JD, Sibenaller ZA, Mapuskar KA, Wagner BA, Cramer-Morales KL, Furqan M et al. O2(-) and H2O2-Mediated Disruption of Fe Metabolism Causes the Differential Susceptibility of NSCLC and GBM Cancer Cells to Pharmacological Ascorbate. Cancer Cell. 2017;31(4):487–500 e488.

  53. Toyokuni S, Okamoto K, Yodoi J, Hiai H. Persistent oxidative stress in cancer. FEBS Lett. 1995;358:1–3.

  54. Ohta T, Iijima K, Miyamoto M, Nakahara I, Tanaka H, Ohtsuji M, et al. Loss of Keap1 function activates Nrf2 and provides advantages for lung cancer cell growth. Cancer Res. 2008;68(5):1303–9.

    Article  CAS  Google Scholar 

  55. Taguchi K, Yamamoto M. The KEAP1-NRF2 System in Cancer. Front Oncol. 2017;7:85.

    Article  Google Scholar 

  56. Motooka Y, Toyokuni S. Ferroptosis as ultimate target of cancer therapy. Antioxid Redox Signal. 2022. https://doi.org/10.1089/ars.2022.0048.

    Article  Google Scholar 

  57. Toyokuni S, Miyake N, Hiai H, Hagiwara M, Kawakishi S, Osawa T, et al. The monoclonal antibody specific for the 4-hydroxy-2-nonenal histidine adduct. FEBS Lett. 1995;359(2–3):189–91.

    Article  CAS  Google Scholar 

  58. Ozeki M, Miyagawa-Hayashino A, Akatsuka S, Shirase T, Lee WH, Uchida K, et al. Susceptibility of actin to modification by 4-hydroxy-2-nonenal. J Chromatogr B Analyt Technol Biomed Life Sci. 2005;827(1):119–26.

    Article  CAS  Google Scholar 

  59. Zheng H, Jiang L, Tsuduki T, Conrad M, Toyokuni S. Embryonal erythropoiesis and aging exploit ferroptosis. Redox Biol. 2021;48: 102175.

    Article  CAS  Google Scholar 

  60. Van Do B, Gouel F, Jonneaux A, Timmerman K, Gele P, Petrault M, et al. Ferroptosis, a newly characterized form of cell death in Parkinson’s disease that is regulated by PKC. Neurobiol Dis. 2016;94:169–78.

    Article  Google Scholar 

  61. Masaldan S, Bush AI, Devos D, Rolland AS, Moreau C. Striking while the iron is hot: Iron metabolism and ferroptosis in neurodegeneration. Free Radic Biol Med. 2019;133:221–33.

    Article  CAS  Google Scholar 

  62. Proneth B, Conrad M. Ferroptosis and necroinflammation, a yet poorly explored link. Cell Death Differ. 2019;26(1):14–24.

    Article  CAS  Google Scholar 

  63. Vogelstein B, Kinzler KW. The genetic basis of human cancer. New York: McGraw-Hill; 1998.

    Google Scholar 

  64. Toyokuni S. Oxidative stress as an iceberg in carcinogenesis and cancer biology. Arch Biochem Biophys. 2016;595:46–9.

    Article  CAS  Google Scholar 

  65. Fearon ER. Human cancer syndromes: clues to the origin and nature of cancer. Science. 1997;278(5340):1043–50.

    Article  CAS  Google Scholar 

  66. Narod SA, Foulkes WD. BRCA1 and BRCA2: 1994 and beyond. Nat Rev Cancer. 2004;4(9):665–76.

    Article  CAS  Google Scholar 

  67. Evans DG, Barwell J, Eccles DM, Collins A, Izatt L, Jacobs C, et al. The Angelina Jolie effect: how high celebrity profile can have a major impact on provision of cancer related services. Breast Cancer Res. 2014;16(5):442.

    Article  Google Scholar 

  68. Momozawa Y, Sasai R, Usui Y, Shiraishi K, Iwasaki Y, Taniyama Y, et al. Expansion of Cancer Risk Profile for BRCA1 and BRCA2 Pathogenic Variants. JAMA Oncol. 2022;8(6):871–8.

    Article  Google Scholar 

  69. Casella D, Di Taranto G, Marcasciano M, Sordi S, Kothari A, Kovacs T, et al. Nipple-sparing bilateral prophylactic mastectomy and immediate reconstruction with TiLoop((R)) Bra mesh in BRCA1/2 mutation carriers: A prospective study of long-term and patient reported outcomes using the BREAST-Q. Breast. 2018;39:8–13.

    Article  CAS  Google Scholar 

  70. Metcalfe K, Eisen A, Senter L, Armel S, Bordeleau L, Meschino WS, et al. International trends in the uptake of cancer risk reduction strategies in women with a BRCA1 or BRCA2 mutation. Br J Cancer. 2019;121(1):15–21.

    Article  Google Scholar 

  71. Miller LD, Coffman LG, Chou JW, Black MA, Bergh J, D’Agostino R Jr, et al. An iron regulatory gene signature predicts outcome in breast cancer. Cancer Res. 2011;71(21):6728–37.

    Article  CAS  Google Scholar 

  72. Torti SV, Manz DH, Paul BT, Blanchette-Farra N, Torti FM. Iron and Cancer. Annu Rev Nutr. 2018;38:97–125.

    Article  CAS  Google Scholar 

  73. Yamaguchi K, Mandai M, Toyokuni S, Hamanishi J, Higuchi T, Takakura K, et al. Contents of endometriotic cysts, especially the high concentration of free iron, are a possible cause of carcinogenesis in the cysts through the iron-induced persistent oxidative stress. Clin Cancer Res. 2008;14(1):32–40.

    Article  CAS  Google Scholar 

  74. Kobayashi H, Yamashita Y, Iwase A, Yoshikawa Y, Yasui H, Kawai Y et al. The ferroimmunomodulatory role of ectopic endometriotic stromal cells in ovarian endometriosis. Fertil Steril. 2012;98(2):415–422 e411–412.

  75. Mori M, Ito F, Shi L, Wang Y, Ishida C, Hattori Y, et al. Ovarian endometriosis-associated stromal cells reveal persistently high affinity for iron. Redox Biol. 2015;6:578–86.

    Article  CAS  Google Scholar 

  76. Kajiyama H, Suzuki S, Yoshihara M, Tamauchi S, Yoshikawa N, Niimi K, et al. Endometriosis and cancer. Free Radic Biol Med. 2019;133:186–92.

    Article  CAS  Google Scholar 

  77. Miki Y, Swensen J, Shattuck-Eidens D, Futreal PA, Harshman K, Tavtigian S, et al. A strong candidate for the breast and ovarian cancer susceptibility gene BRCA1. Science. 1994;266(5182):66–71.

    Article  CAS  Google Scholar 

  78. Evers B, Jonkers J. Mouse models of BRCA1 and BRCA2 deficiency: past lessons, current understanding and future prospects. Oncogene. 2006;25(43):5885–97.

    Article  CAS  Google Scholar 

  79. Liu X, Holstege H, van der Gulden H, Treur-Mulder M, Zevenhoven J, Velds A, et al. Somatic loss of BRCA1 and p53 in mice induces mammary tumors with features of human BRCA1-mutated basal-like breast cancer. Proc Natl Acad Sci U S A. 2007;104(29):12111–6.

    Article  CAS  Google Scholar 

  80. Nakamura Y, Kubota J, Nishimura Y, Nagata K, Nishimura M, Daino K, et al. Brca 1(L63X) (/+) rat is a novel model of human BRCA1 deficiency displaying susceptibility to radiation-induced mammary cancer. Cancer Sci. 2022;113(10):3362–75.

    Article  CAS  Google Scholar 

  81. Akatsuka S, Li GH, Toyokuni S. Superiority of rat over murine model for studies on the evolution of cancer genome. Free Radic Res. 2018;52(11–12):1323–7.

    Article  CAS  Google Scholar 

  82. Okada S, Midorikawa O. Induction of rat renal adenocarcinoma by Fe-nitrilotriacetate (Fe-NTA). Jpn Arch Intern Med. 1982;29:485–91.

    CAS  Google Scholar 

  83. Kong Y, Akatsuka S, Motooka Y, Zheng H, Cheng Z, Shiraki Y, et al. BRCA1 haploinsufficiency promotes chromosomal amplification under Fenton reaction-based carcinogenesis through ferroptosis-resistance. Redox Biol. 2022;54: 102356.

    Article  CAS  Google Scholar 

  84. Cheng Z, Akatsuka S, Li GH, Mori K, Takahashi T, Toyokuni S. Ferroptosis resistance determines high susceptibility of murine A/J strain to iron-induced renal carcinogenesis. Cancer Sci. 2022;113(1):65–78.

    Article  CAS  Google Scholar 

  85. Li GH, Akatsuka S, Chew SH, Jiang L, Nishiyama T, Sakamoto A, et al. Fenton reaction-induced renal carcinogenesis in Mutyh-deficient mice exhibits less chromosomal aberrations than the rat model. Pathol Int. 2017;67(11):564–74.

    Article  CAS  Google Scholar 

  86. Tanaka T, Iwasa Y, Kondo S, Hiai H, Toyokuni S. 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. 1999;18:3793–7.

    Article  CAS  Google Scholar 

  87. Inagaki-Kawata Y, Yoshida K, Kawaguchi-Sakita N, Kawashima M, Nishimura T, Senda N, et al. Genetic and clinical landscape of breast cancers with germline BRCA1/2 variants. Commun Biol. 2020;3(1):578.

    Article  CAS  Google Scholar 

  88. Chen Y, Olopade OI. MYC in breast tumor progression. Expert Rev Anticancer Ther. 2008;8(10):1689–98.

    Article  CAS  Google Scholar 

  89. Grushko TA, Dignam JJ, Das S, Blackwood AM, Perou CM, Ridderstrale KK, et al. MYC is amplified in BRCA1-associated breast cancers. Clin Cancer Res. 2004;10(2):499–507.

    Article  CAS  Google Scholar 

  90. Luo Y, Akatsuka S, Motooka Y, Kong Y, Zheng H, Mashimo T, et al. BRCA1 haploinsufficiency impairs iron metabolism to promote chrysotile-induced mesothelioma via ferroptosis-resistance. Cancer Sci. 2022. https://doi.org/10.1111/cas.15705.

    Article  Google Scholar 

Download references

Acknowledgements

The author (YK) would like to take this opportunity to thank the “Interdisciplinary Frontier Next-Generation Researcher Program of the 10 Tokai Higher Education and Research System.” The authors thank Division for Medical Research Engineering, Nagoya University Graduate School of Medicine for technical assistance.

Conflict of interest

None.

Funding

This work was supported, in part, by JST CREST (Grant Number JPMJCR19H4) and JSPS Kakenhi (Grant Number JP19H05462 and JP20H05502) to ST. This work was financially supported by JST SPRING, Grant Number JPMJSP2125 to YK.

Author information

Authors and Affiliations

Authors

Contributions

ST, YK, YM and SA conceived, wrote and organized the manuscript, prepared the figures, and contributed to the discussion. The author(s) read and approved the final manuscript.

Corresponding author

Correspondence to Shinya Toyokuni.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

All the authors agreed to an author of this review article.

Competing interests

All the authors declare no conflict of interest to present.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Toyokuni, S., Kong, Y., Motooka, Y. et al. Environmental impact on carcinogenesis under BRCA1 haploinsufficiency. Genes and Environ 45, 2 (2023). https://doi.org/10.1186/s41021-023-00258-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s41021-023-00258-5