Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2020 Mar 26.
Published in final edited form as: Biol Trace Elem Res. 2015 Mar 22;166(1):34–40. doi: 10.1007/s12011-015-0306-7

Disruption of Mitotic Progression by Arsenic

J Christopher States 1
PMCID: PMC7098731  NIHMSID: NIHMS1567674  PMID: 25796515

Abstract

Arsenic is an enigmatic xenobiotic that causes a multitude of chronic diseases including cancer and also is a therapeutic with promise in cancer treatment. Arsenic causes mitotic delay and induces aneuploidy in diploid human cells. In contrast, arsenic causes mitotic arrest followed by an apoptotic death in a multitude of virally transformed cells and cancer cells. We have explored the hypothesis that these differential effects of arsenic exposure are related by arsenic disruption of mitosis and are differentiated by the target cell’s ability to regulate or modify cell cycle checkpoints. Functional p53/CDKN1A axis has been shown to mitigate the mitotic block and to be essential to induction of aneuploidy. More recent preliminary data suggest that microRNA modulation of chromatid cohesion also may play a role in escape from mitotic block and in generation of chromosomal instability. Other recent studies suggest that arsenic may be useful in treatment of solid tumors when used in combination with other cytotoxic agents such as cisplatin.


Arsenic is an enigmatic xenobiotic that on one hand causes a variety of chronic diseases and on the other has therapeutic uses. Chronic arsenic exposure, most commonly as arsenite or arsenate contaminating drinking water, is a worldwide problem affecting 140 million people [1, 2]. Chronic arsenic exposure causes cancer, cardiovascular disease, neurological disorders, reproductive problems, chronic pulmonary disease, ocular disorders, pigmentary changes, and diabetes [38]. Arsenicals have been used in Chinese traditional medicines for centuries [9], are still used to treat infectious tropical diseases [10], and arsenic trioxide (ATO, As2O3) is approved by USFDA to treat acute promyelocytic leukemia (APL) [11]. This review focuses on the role that arsenic-induced mitotic disruption plays in carcinogenesis and potentially in chemotherapy of solid tumors.

Arsenic is a group I human carcinogen, and chronic inorganic arsenic exposure causes skin, lung, and bladder cancer and likely causes prostate, liver, and kidney cancer in humans [12]. Transplacental inorganic arsenic exposure causes lung, liver, ovary, and adrenal tumors in mice [13, 14]. Proposed modes of action in arsenic carcinogenesis include DNA repair inhibition, co-mutagenicity, altered DNA methylation, oxidative stress, cell proliferation, and aneuploidy, including both structural and numerical chromosomal abnormalities. There are ample data supporting each of these proposed modes, but aneuploidy was deemed the most plausible mechanism or mode of action for arsenic carcinogenicity [15, 16]. Since these NRC reports were published, much research has been performed in pursuit of modes other than aneuploidy. However, appreciation of the role of aneuploidy, or chromosomal instability, as a major driver of carcinogenesis is regaining attention [1719]. Thus, this review is focused on mechanisms related to aneuploidy. The disruption of mitosis by arsenicals is central to the induction of aneuploidy.

Aneuploidy classically refers to chromosomal numerical abnormalities, e.g., trisomy 21 or a loss of heterozygosity event. Numerical chromosomal abnormalities arise from chromatid mis-segregation events resulting in uneven distribution of chromatids in daughter cells [19]. Structural chromosomal abnormalities arise from clastogenic events. These structural abnormalities include translocations, partial loss of chromatids, gene amplifications, and copy number variations, all common events in carcinogenesis [19].

ATO is an effective chemotherapeutic in combination with all trans retinoic acid (ATRA) for treating acute APL [2022]. The mechanism of action for ATO in treating APL appears to be by enhancing SUMOylation and subsequent degradation of the PML-RAR fusion protein (result of a translocation) that drives this malignancy [23]. There has been interest in the potential use of ATO in treatment of solid tumors, but clinical trials of ATO as a single agent have failed. However, clinical trials of ATO in combination chemotherapies have not been performed.

Many investigators have used sodium arsenite rather than ATO for in vitro studies. Chemically, these agents are both converted to arsenous acid and exist as arsenous acid [As(OH)3] in neutral solution [24]. Indeed, Trisenox®, the pharmacological formulation of ATO used in the clinic to treat APL, is prepared by dissolution of ATO in 30 mM NaOH followed by titration with HCl to near neutral pH (http://www.trisenox.com/hcp/trisenox-prescribing-information.pdf). ATO and sodium arsenite induce apoptosis in vitro in many types of cancer cells derived from solid tumors including melanoma [25], lung cancer [26], neuroblastoma [27], ovarian cancer [28, 29], transitional cell carcinoma [30], and prostate cancer [29]. Arsenite has long been known to disrupt mitosis, but normal diploid cells can survive this disruption albeit with an increase in aneuploidy. Normal diploid fibroblasts incubated with 5 μM sodium arsenite exhibited a prolonged mitotic delay from which the cells recovered with an increased incidence of aneuploidy [31]. The toxic effect of arsenic clearly is related to the effects on mitotic progression. Confluent telomerase immortalized diploid human fibroblasts, which retain most characteristics of their non-immortalized parental cells [32], tolerate up to 30 μM NaAsO2 for at least 6 days with no overt signs of toxicity whereas growing cells are dying by 48 h [33]. Incubation of phytohemaglutinin-stimulated peripheral blood lymphocytes with sodium arsenite in vitro induced mitotic arrest and aneuploidy with arsenite concentrations as low as 100 fM. [34]. Mitotic index and aneuploidy showed concentration response in these lymphocytes from 100 fM. to 10 nM arsenite. Aneuploidy also is induced in telomerase-immortalized diploid human fibroblasts exposed to modest concentrations of arsenite [35]. SV40-transfomed human fibroblasts arrest in mitosis and undergo an apoptotic death when exposed to 1–5 μM sodium arsenite [33]. Thus, the induction of aneuploidy is dependent on the ability to escape the arsenite-induced inhibition of mitotic progression.

Cell cycle control mechanisms play an important role in cell survival after arsenic exposure. We showed that SV40-transformed human skin fibroblasts were sensitive to arsenic-induced mitotic arrest and underwent apoptotic death [33]. SV40 transformation results in sequestration of p53 and retinoblastoma (Rb) proteins by the SV40 T-antigen. This sequestration abrogates the ability of p53 and Rb to perform their cell cycle arrest functions. The role of p53 in escaping arsenic-induced mitotic arrest and avoiding the ensuing mitotic death was demonstrated in TR9–7 cells, a human fibroblast cell line with a tetracycline-regulated p53 expression cassette [36]. The induction of cyclin-dependent kinase inhibitor 1A (CDKN1A, aka p21CIP1/WAF1) was shown to mediate the p53 effect [37]. Suppression of CDKN1A induction by transfected small interfering RNA (siRNA) directed to CDKN1A messenger RNAs (mRNAs) caused arsenite-treated p53 expressing cells to undergo mitotic death at high levels similar to cells not expressing p53. Thus, consistent with roles for p53 and CDKN1A in escape from arsenic-induced mitotic arrest, these proteins are also necessary for the induction of aneuploidy in telomerase-immortalized diploid human fibroblasts [35]. Arsenic exposure has long been known to induce micronuclei which can contain either whole chromosomes or fragments of chromosomes [3842]. Micronuclei containing whole chromosomes are demonstrated by in situ hybridization with a centromeric probe and indicate a classic aneuploidogenic event giving rise to chromosomal numerical abnormality. Centromere negative micronuclei indicate a chromosomal fragment from a clastogenic event that gave rise to this chromosomal structural abnormality. Suppression of p53 or CDKN1A by transfection of siRNAs targeting p53 or CDKN1A mRNAs caused a shift from a predominance of centromere positive to negative micronuclei indicating a shift from aneuploidogenesis to clastogenesis [35]. Thus, these proteins that have central roles in mediating cell cycle checkpoint responses are essential to an imperfect escape from mitotic arrest resulting in aneuploidy.

A wide variety of cancer cell lines including melanoma, HeLa, glioma, and lung, ovarian and breast cancer cells undergo an apoptotic death after mitotic arrest induced by ATO or sodium arsenite exposure [4347]. This apoptotic death follows centrosome fragmentation [48, 49] and can be prevented by induction of the cyclin-dependent kinase inhibitor CDKN1A (aka p21CIP1/WAF1) in p53 expressing cells [37]. It is likely that induction of CDKN1A serves to inhibit cyclin-dependent kinase 1 (CDK1), thus releasing the cells from the CDK1-dependent mitotic arrest [25]. This conclusion is supported by the observation that roscovitine, a CDK inhibitor, also releases cells from arsenite-induced mitotic arrest [25]. The centrosome fragmentation can be augmented by heat shock or inhibition of the heat shock system [50, 49]. Thus, the ability to induce mitotic arrest and apoptosis also may play a role in chemotherapy of solid tumors. However, recent studies suggest that arsenicals may have greater potential acting in combination with other chemotherapeutics such as cisplatin [5159]. Detailed studies in ovarian cancer models suggest that arsenite enhances sensitivity to cisplatin but only in cells expressing p53 [57]. Both in cells in vitro [57] and in xenotransplant tumors in mice [58], arsenite co-treatment prevents the responses of DNA repair systems associated with cisplatin resistance. Induction of nucleotide excision repair genes and suppression of mismatch repair genes by cisplatin are inhibited by the arsenite co-treatment. Further investigation of effects on cell cycle revealed that arsenite co-treatment resulted in abrogration of the G2 checkpoint normally activated by cisplatin; the cells then passed mitosis without dividing and entered into a pseudo-G1 state where they underwent apoptosis [59]. These results point to a complex effect of arsenite on mitotic progression that is not yet completely understood.

How arsenic, which inhibits mitosis, can induce cancer which is a disease of uncontrolled cell replication appears paradoxical on its surface. The mechanisms of arsenic disruption of mitosis likely play a role both in carcinogenesis and in induction of mitotic death. The in vitro studies discussed above implicate p53 and CDKN1A in escape from arsenic-induced mitotic arrest and subsequent aneuploidy. Failure to escape the mitotic arrest can lead to mitotic death, also referred to as mitotic catastrophe in earlier publications. This induction of mitotic death, consequent to failed cytokinesis, is a major mechanism of apoptotic death induced by arsenicals in many types of cancer cells. Thus, we believe that the response to arsenical-induced mitotic arrest is the common link between arsenical-induced carcinogenesis in diploid cells and arsenical-induced apoptosis in cancer cells (Fig. 1).

Fig. 1.

Fig. 1

Cell checkpoint control mechanisms mediate the carcinogenic (aneuploidogenic) vs chemotherapeutic (apoptotic) response to arsenic exposure. Lack of p53 is associated with failed cytokinesis and induction of mitotic death. Functional p53/CDKN1A (p21) axis relieves mitotic block, and miR-186 overexpression allows anaphase progression and contributes to aneuploidy by promoting premature chromatid separation

The mitotic catastrophe we observed in arsenite-treated cells included degradation of the chromosomes. The appearance of these catastrophic chromosome spreads was very similar to the chromosome fragmentation observed by others in cancer cells treated with a variety of stressors [6062]. Chromosome fragmentation can lead to genome chaos [63], a severe form of aneuploidy, and contribute to carcinogenesis. Arsenite clearly induces cellular stress and can induce centrosome fragmentation [49] which can contribute to chromosome fragmentation [60]. However, in our studies, the end result of the mitotic disruption was cell death with activated caspases [44, 25, 37] clearly indicating that an apoptotic death pathway is activated. Indeed, the catastrophic mitotic figures are absent, and the morphologically distinct mitotic figures are more abundant when caspases are inhibited [25, 37]. Thus, at least at concentrations relevant to those achieved in chemotherapy, it is unlikely that the arsenite-induced mitotic death will contribute to genome chaos. The impact of low, environmental concentrations remains to be investigated.

Progression through mitosis is a complicated process and involves four morphologically distinct phases. Cell cycle transitions are governed by CDKs. Most transitions require induction of cyclin expression, but progression from mitosis (M phase) to G1 phase is different. Loss of cyclin B is required for this transition. Cyclin B is the cognate cyclin for CDK1, the master kinase governing entry into and progression through the early stages of mitosis. Thus, normal expression of cyclin B in G2 and early M activates CDK1 which is essential for progression through G2 and prophase to meta-phase. Cells are held at the metaphase/anaphase junction by the spindle assembly checkpoint until all chromosomes are attached to the spindle. A functional spindle assembly checkpoint is necessary for arsenite-induced mitotic arrest [25]. In order to activate the mitotic exit process and to progress through anaphase to telophase, cyclin B normally is ubiquitinylated by the anaphase promoting complex/cyclosome (APC/C) and degraded by the proteasome to inactivate CDK1 (Fig. 2) [64]. Cyclin B is stabilized in arsenite-exposed cells [37, 25] suggesting inhibition of APC/C. Inhibition of CDK1 by CDKN1A [37] or roscovitine [25] allows mitotic exit in arsenite-arrested cells indicating that persistent CDK1 activity is preventing mitotic exit. The p53 induction of CDKN1A in arsenite-exposed cells, as discussed above, appears to be a mechanism for overcoming this phase of the mitotic block in diploid cells.

Fig. 2.

Fig. 2

Arsenic impact on mitotic progression. Progression from prophase through telophase to aneuploid progeny is diagrammed across the middle of the figure. Key events in the transitions are driven by cyclin B/CDK1 activity (prophase to metaphase progression) or inactivation (mitotic exit: metaphase to anaphase transition) and by separase activation and cleavage of cohesins to allow chromatid separation in anaphase. Cyclin B and securin are ubiquitinylated by the anaphase promoting complex/cyclosome (APC/C) and degraded by the proteasome. Cyclin B and securin are stabilized by arsenic (As) exposure. Arsenic induces p53 expression which, in turn, induces CDKN1A that then inhibits CDK1 activity and allows mitotic exit. miR-186 is overexpressed in arsenic-induced aquamous cell carcinoma and is known to suppress securin expression. It is hypothesized that the lower levels of securin would allow separase activation in the presence of arsenic and also would contribute to aneuploidy by inducing premature chromatid separation

Anaphase is the period in which the chromatids separate and move toward the spindle poles. The chromatids are held together in the chromosomes by a protein complex of cohesins. In order to separate the chromatids, the cohesins must be cleaved by the protease separase. Separase is held in an inactive state by binding of securin until mitotic exit is activated [65]. Then, securin is ubiquitinylated by the APC/C and degraded by the proteasome, thus releasing separase to cleave the cohesins in the metaphase to anaphase transition (Fig. 2) [64]. Securin also is stabilized by arsenite [25], also consistent with inhibition of the APC/C. Until recently, it was not clear how cells might overcome the stabilization of securin. MicroRNA expression was compared in biopsies of hyperkeratoses and squamous cell carcinomas obtained from West Bengali patients exposed to high levels of arsenic in their drinking water. The hyperkeratoses are premalignant lesions that give rise to both basal and squamous cell carcinomas and are pathognomonic for arsenicosis. Total RNA was purified from keratinocytes laser captured microdissected from the skin lesions, and microRNA expression was profiled on qRT-PCR arrays. The microRNA most elevated in squamous cell carcinoma relative to hyperkeratosis is hsa-miR-186 (unpublished data). Securin (aka PTTG1) is a validated target of this microRNA [66]. Thus, we propose that elevated expression of hsa-miR-186 in keratinocytes chronically exposed to arsenic provides the suppression of securin needed to overcome the arsenic exposure-induced mitotic disruption and to progress to a malignant state. In addition, overexpression of hsa-miR-186 would likely induce a state of chromosomal instability due to premature separation of chromatids further contributing to carcinogenesis. We are currently testing this hypothesis.

Summary

The studies discussed clearly indicate that arsenite at physiological levels (10 fM–10 nM) and therapeutic levels (1–10 μM) disrupt normal mitotic progression. The consequences of this disruption depend upon the ability of the cell to regulate cell cycle checkpoints. Clearly, cyclin B and securin are stabilized by arsenite. The cyclin B stabilization results in persistent CDK1 activity which prevents mitotic exit. Cells with an intact p53/CDKN1A axis induce CDKN1A which, in turn, inhibits CDK1, thus activating mitotic exit. However, securin also is stabilized in arsenite-exposed cells. Recent preliminary results suggest that miR-186 is induced by chronic arsenic exposure and this microRNA suppresses securin expression. This suppression would lead not only to facilitated progression from metaphase through anaphase but also to chromosomal instability. The stabilization of cyclin B and securin suggests that arsenite is inhibiting the APC/C. Inhibition of the APC/C combined with microRNA suppression of securin is a likely mode of action for arsenic carcinogenesis by induction of aneuploidy. In cells lacking adequate cell cycle checkpoint control, such as most cancer cells, arsenite induces mitotic arrest leading to centrosome fragmentation and mitotic death. In cells co-treated with cytotoxic drugs like cisplatin, the mitotic disruption causes entry into a pseudo-G1 state where the cells undergo apoptosis. These findings suggest that ATO has potential use in combination chemotherapy for cancer.

Acknowledgments

The author is indebted to the students and collaborators who contributed to the work discussed in this review. In no particular order, these include Sam McNeely, Frazier Taylor, Ana Maria Salazar, Ashok Giri, Vanessa States, Heather Miller, Clarisse Muenyi, Josh Masters, and Patricia Ostroskey-Wegman. The work discussed was supported in part by USPHS grant ES011314.

References

  • 1.Mondal D, Banerjee M, Kundu M, Banerjee N, Bhattacharya U, Giri AK, Ganguli B, Sen Roy S, Polya DA (2010) Comparison of drinking water, raw rice and cooking of rice as arsenic exposure routes in three contrasting areas of West Bengal, India. Environ Geochem Health 32(6):463–477. doi: 10.1007/s10653-010-9319-5 [DOI] [PubMed] [Google Scholar]
  • 2.Polya D, Charlet L (2009) Environmental science: rising arsenic risk? Nat Geosci 2(6):383–384 [Google Scholar]
  • 3.Yoshida T, Yamauchi H, Fan Sun G (2004) Chronic health effects in people exposed to arsenic via the drinking water: dose-response relationships in review. Toxicol Appl Pharmacol 198(3):243–252. doi: 10.1016/j.taap.2003.10.022 [DOI] [PubMed] [Google Scholar]
  • 4.Kapaj S, Peterson H, Liber K, Bhattacharya P (2006) Human health effects from chronic arsenic poisoning—a review. J Environ Sci Health, Part A: Tox Hazard Subst Environ Eng 41(10):2399–2428. doi: 10.1080/10934520600873571 [DOI] [PubMed] [Google Scholar]
  • 5.Vahidnia A, van der Voet GB, de Wolff FA (2007) Arsenic neurotoxicity—a review. Hum Exp Toxicol 26(10):823–832. doi: 10.1177/0960327107084539 [DOI] [PubMed] [Google Scholar]
  • 6.Guha Mazumder DN (2008) Chronic arsenic toxicity & human health. Indian J Med Res 128(4):436–447 [PubMed] [Google Scholar]
  • 7.Soto-Martinez M, Sly PD (2010) Relationship between environmental exposures in children and adult lung disease: the case for outdoor exposures. Chron Respir Dis 7(3):173–186. doi: 10.1177/1479972309345929 [DOI] [PubMed] [Google Scholar]
  • 8.Guha Mazumder D, Dasgupta UB (2011) Chronic arsenic toxicity: studies in West Bengal, India. Kaohsiung J Med Sci 27(9):360–370. doi: 10.1016/j.kjms.2011.05.003 [DOI] [PubMed] [Google Scholar]
  • 9.Liu J, Lu Y, Wu Q, Goyer RA, Waalkes MP (2008) Mineral arsenicals in traditional medicines: orpiment, realgar, and arsenolite. J Pharmacol Exp Ther 326(2):363–368. doi: 10.1124/jpet.108.139543 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Kennedy PG (2013) Clinical features, diagnosis, and treatment of human African trypanosomiasis (sleeping sickness). Lancet Neurol 12(2):186–194. doi: 10.1016/S1474-4422(12)70296-X [DOI] [PubMed] [Google Scholar]
  • 11.Antman KH (2001) Introduction: the history of arsenic trioxide in cancer therapy. Oncologist 6(Suppl 2):1–2 [DOI] [PubMed] [Google Scholar]
  • 12.Humans. IWGotEoCRt (2012) Arsenic, metals, fibres, and dusts. IARC Monogr Eval Carcinog Risks Hum 100(Pt C):11–465 [PMC free article] [PubMed] [Google Scholar]
  • 13.Waalkes MP, Liu J, Diwan BA (2007) Transplacental arsenic carcinogenesis in mice. Toxicol Appl Pharmacol 222(3):271–280. doi: 10.1016/j.taap.2006.12.034 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Tokar EJ, Diwan BA, Ward JM, Delker DA, Waalkes MP (2011) Carcinogenic effects of “whole-life” exposure to inorganic arsenic in CD1 mice. Toxicol Sci 119(1):73–83. doi: 10.1093/toxsci/kfq315 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Subcommittee on Arsenic in Drinking Water, National Research Council (1999) Arsenic in drinking water. National Academy Press, Washington [Google Scholar]
  • 16.Subcommittee to Update the 1999 Arsenic in Drinking Water Report, Committee on Toxicology, Board on Environmental Studies and Toxicology, National Research Council (2001) Arsenic in drinking water: 2001 update. National Academy Press, Washington [Google Scholar]
  • 17.Cassidy LD, Liau SS, Venkitaraman AR (2014) Chromosome instability and carcinogenesis: insights from murine models of human pancreatic cancer associated with BRCA2 inactivation. Mol Oncol 8(2):161–168. doi: 10.1016/j.molonc.2013.10.005 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Wu Y, Antony S, Meitzler JL, Doroshow JH (2014) Molecular mechanisms underlying chronic inflammation-associated cancers. Cancer Lett 345(2):164–173. doi: 10.1016/j.canlet.2013.08.014 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Kops GJPL, Weaver BAA, Cleveland DW (2005) On the road to cancer: aneuploidy and the mitotic checkpoint. Nat Rev Cancer 5(10):773–785 [DOI] [PubMed] [Google Scholar]
  • 20.Shen ZX, Chen GQ, Ni JH, Li XS, Xiong SM, Qiu QY, Zhu J, Tang W, Sun GL, Yang KQ, Chen Y, Zhou L, Fang ZW, Wang YT, Ma J, Zhang P, Zhang TD, Chen SJ, Chen Z, Wang ZY (1997) Use of arsenic trioxide (As2O3) in the treatment of acute promyelocytic leukemia (APL): II. Clinical efficacy and pharmacokinetics in relapsed patients. Blood 89(9):3354–3360 [PubMed] [Google Scholar]
  • 21.Iland HJ, Seymour JF (2013) Role of arsenic trioxide in acute promyelocytic leukemia. Curr Treat Options Oncol 14(2):170–184. doi: 10.1007/s11864-012-0223-3 [DOI] [PubMed] [Google Scholar]
  • 22.Lo-Coco F, Avvisati G, Vignetti M, Thiede C, Orlando SM, Iacobelli S, Ferrara F, Fazi P, Cicconi L, Di BE, Specchia G, Sica S, Divona M, Levis A, Fiedler W, Cerqui E, Breccia M, Fioritoni G, Salih HR, Cazzola M, Melillo L, Carella AM, Brandts CH, Morra E, von Lilienfeld-Toal M, Hertenstein B, Wattad M, Lubbert M, Hanel M, Schmitz N, Link H, Kropp MG, Rambaldi A, La NG, Luppi M, Ciceri F, Finizio O, Venditti A, Fabbiano F, Dohner K, Sauer M, Ganser A, Amadori S, Mandelli F, Dohner H, Ehninger G, Schlenk RF, Platzbecker U (2013) Retinoic acid and arsenic trioxide for acute promyelocytic leukemia. N Engl J Med 369(2): 111–121. doi: 10.1056/NEJMoa1300874 [DOI] [PubMed] [Google Scholar]
  • 23.Zhang XW, Yan XJ, Zhou ZR, Yang FF, Wu ZY, Sun HB, Liang WX, Song AX, Lallemand-Breitenbach V, Jeanne M, Zhang QY, Yang HY, Huang QH, Zhou GB, Tong JH, Zhang Y, Wu JH, Hu HY, de Thé H, Chen SJ, Chen Z (2010) Arsenic trioxide controls the fate of the PML-RARalpha oncoprotein by directly binding PML. Science 328(5975):240–243. doi: 10.1126/science.1183424 [DOI] [PubMed] [Google Scholar]
  • 24.Ramirez-Solis A, Mukopadhyay R, Rosen BP, Stemmler TL (2004) Experimental and theoretical characterization of arsenite in water: insights into the coordination environment of As-O. Inorg Chem 43(9):2954–2959. doi: 10.1021/ic0351592 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.McNeely SC, Taylor BF, States JC (2008) Mitotic arrest-associated apoptosis induced by sodium arsenite in A375 melanoma cells is BUBR1-dependent. Toxicol Appl Pharmacol 231(1):61–67. doi: 10.1016/j.taap.2008.03.020 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Qu GP, Xiu QY, Li B, Liu YA, Zhang LZ (2009) Arsenic trioxide inhibits the growth of human lung cancer cell lines via cell cycle arrest and induction of apoptosis at both normoxia and hypoxia. Toxicol Ind Health 25(8): 505–515. doi: 10.1177/0748233709345936 [DOI] [PubMed] [Google Scholar]
  • 27.Akao Y, Yamada H, Nakagawa Y (2000) Arsenic-induced apoptosis in malignant cells in vitro. Leuk Lymphoma 37(1–2):53–63. doi: 10.3109/10428190009057628 [DOI] [PubMed] [Google Scholar]
  • 28.Kong B, Huang S, Wang W, Ma D, Qu X, Jiang J, Yang X, Zhang Y, Wang B, Cui B, Yang Q (2005) Arsenic trioxide induces apoptosis in cisplatin-sensitive and -resistant ovarian cancer cell lines. IntJ Gynecol Cancer 15(5):872–877. doi: 10.1111/j.1525-1438.2005.00251.x [DOI] [PubMed] [Google Scholar]
  • 29.Uslu R, Sanli UA, Sezgin C, Karabulut B, Terzioglu E, Omay SB, Goker E (2000) Arsenic trioxide-mediated cytotoxicity and apoptosis in prostate and ovarian carcinoma cell lines. Clin Cancer Res 6(12):4957–4964 [PubMed] [Google Scholar]
  • 30.Pu YS, Hour TC, Chen J, Huang CY, Guan JY, Lu SH (2002) Arsenic trioxide as a novel anticancer agent against human transitional carcinoma—characterizing its apoptotic pathway. Anti-Cancer Drugs 13(3):293–300 [DOI] [PubMed] [Google Scholar]
  • 31.Yih LH, Ho IC, Lee TC (1997) Sodium arsenite disturbs mitosis and induces chromosome loss in human fibroblasts. Cancer Res 57(22):5051–5059 [PubMed] [Google Scholar]
  • 32.Porter PC, Clark DR, McDaniel LD, McGregor WG, States JC (2006) Telomerase-immortalized human fibroblasts retain UV-induced mutagenesis and p53-mediated DNA damage responses. DNA Repair 5(1):61–70. doi: 10.1016/j.dnarep.2005.07.005 [DOI] [PubMed] [Google Scholar]
  • 33.States JC, Reiners JJ Jr, Pounds JG, Kaplan DJ, Beauerle BD, McNeely SC, Mathieu P, McCabe MJ Jr (2002) Arsenite disrupts mitosis and induces apoptosis in SV40-transformed human skin fibroblasts. Toxicol Appl Pharmacol 180(2):83–91. doi: 10.1006/taap.2002.9376 [DOI] [PubMed] [Google Scholar]
  • 34.Vega L, Gonsebatt ME, Ostrosky-Wegman P (1995) Aneugenic effect of sodium arsenite on human lymphocytes in vitro: an individual susceptibility effect detected. Mutat Res 334(3):365–373. doi: 10.1016/0165-1161(95)90074-8 [DOI] [PubMed] [Google Scholar]
  • 35.Salazar AM, Miller HL, McNeely SC, Sordo M, Ostrosky-Wegman P, States JC (2010) Suppression of p53 and p21CIP1/WAF1 reduces arsenite-induced aneuploidy. Chem Res Toxicol 23(2):357–364. doi: 10.1021/tx900353v [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.McNeely SC, Xu X, Taylor BF, Zacharias W, McCabe MJ Jr, States JC (2006) Exit from arsenite-induced mitotic arrest is p53 dependent. Environ Health Perspect 114(9):1401–1406 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Taylor BF, McNeely SC, Miller HL, Lehmann GM, McCabe MJ, States JC (2006) p53 suppression of arsenite-induced mitotic catastrophe is mediated by p21CIP1/WAF1. J Pharmacol Exp Ther 318(1):142–151. doi: 10.1124/jpet.106.103077 [DOI] [PubMed] [Google Scholar]
  • 38.Ghosh P, Basu A, Singh KK, Giri AK (2008) Evaluation of cell types for assessment of cytogenetic damage in arsenic exposed population. Mol Cancer 7:45. doi: 10.1186/1476-4598-7-45 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Basu A, Ghosh P, Das JK, Banerjee A, Ray K, Giri AK (2004) Micronuclei as biomarkers of carcinogen exposure in populations exposed to arsenic through drinking water in West Bengal, India: a comparative study in three cell types. Cancer Epidemiol Biomark Prev 13(5):820–827 [PubMed] [Google Scholar]
  • 40.Basu A, Mahata J, Roy AK, Sarkar JN, Poddar G, Nandy AK, Sarkar PK, Dutta PK, Banerjee A, Das M, Ray K, Roychaudhury S, Natarajan AT, Nilsson R, Giri AK (2002) Enhanced frequency of micronuclei in individuals exposed to arsenic through drinking water in West Bengal, India. Mutat Res 516(1–2):29–40 [DOI] [PubMed] [Google Scholar]
  • 41.Ghosh P, Basu A, Mahata J, Basu S, Sengupta M, Das JK, Mukherjee A, Sarkar AK, Mondal L, Ray K, Giri AK (2006) Cytogenetic damage and genetic variants in the individuals susceptible to arsenic-induced cancer through drinking water. Int J Cancer 118(10):2470–2478. doi: 10.1002/ijc.21640 [DOI] [PubMed] [Google Scholar]
  • 42.Gonsebatt ME, Vega L, Salazar AM, Montero R, Guzman P, Blas J, Del Razo LM, Garcia-Vargas G, Albores A, Cebrian ME, Kelsh M, Ostrosky-Wegman P (1997) Cytogenetic effects in human exposure to arsenic. Mutat Res 386(3):219–228 [DOI] [PubMed] [Google Scholar]
  • 43.Ling YH, Jiang JD, Holland JF, Perez-Soler R (2002) Arsenic trioxide produces polymerization of microtubules and mitotic arrest before apoptosis in human tumor cell lines. Mol Pharmacol 62(3): 529–538 [DOI] [PubMed] [Google Scholar]
  • 44.McNeely SC, Belshoff AC, Taylor BF, Fan TW, McCabe MJ Jr, Pinhas AR, States JC (2008) Sensitivity to sodium arsenite in human melanoma cells depends upon susceptibility to arsenite-induced mitotic arrest. Toxicol Appl Pharmacol 229(2):252–261. doi: 10.1016/j.taap.2008.01.020 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Wu YC, Yen WY, Yih LH (2008) Requirement of a functional spindle checkpoint for arsenite-induced apoptosis. J Cell Biochem 105(3):678–687. doi: 10.1002/jcb.21861 [DOI] [PubMed] [Google Scholar]
  • 46.Yih LH, Wu YC, Hsu NC, Kuo HH (2012) Arsenic trioxide induces abnormal mitotic spindles through a PIP4KIIgamma/Rho pathway. Toxicol Sci 128(1):115–125. doi: 10.1093/toxsci/kfs129 [DOI] [PubMed] [Google Scholar]
  • 47.Yih LH, Hsueh SW, Luu WS, Chiu TH, Lee TC (2005) Arsenite induces prominent mitotic arrest via inhibition of G2 checkpoint activation in CGL-2 cells. Carcinogenesis 26(1):53–63. doi: 10.1093/carcin/bgh295 [DOI] [PubMed] [Google Scholar]
  • 48.Yih LH, Tseng YY, Wu YC, Lee TC (2006) Induction of centrosome amplification during arsenite-induced mitotic arrest in CGL-2 cells. Cancer Res 66(4):2098–2106. doi: 10.1158/0008-5472.CAN-05-2308 [DOI] [PubMed] [Google Scholar]
  • 49.Taylor BF, McNeely SC, Miller HL, States JC (2008) Arsenite-induced mitotic death involves stress response and is independent of tubulin polymerization. Toxicol Appl Pharmacol 230(2):235–246. doi: 10.1016/j.taap.2008.02.030 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Wu YC, Yen WY, Lee TC, Yih LH (2009) Heat shock protein inhibitors, 17-DMAG and KNK437, enhance arsenic trioxide-induced mitotic apoptosis. Toxicol Appl Pharmacol 236(2):231–238. doi: 10.1016/j.taap.2009.02.003 [DOI] [PubMed] [Google Scholar]
  • 51.Helm CW, States JC (2009) Enhancing the efficacy of cisplatin in ovarian cancer treatment—could arsenic have a role. J Ovarian Res 2:2. doi: 10.1186/1757-2215-2-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Nakaoka T, Ota A, Ono T, Karnan S, Konishi H, Furuhashi A, Ohmura Y, Yamada Y, Hosokawa Y, Kazaoka Y (2014) Combined arsenic trioxide-cisplatin treatment enhances apoptosis in oral squamous cell carcinoma cells. Cell Oncol (Dordr) 37(2): 119–129. doi: 10.1007/s13402-014-0167-7 [DOI] [PubMed] [Google Scholar]
  • 53.Byun JM, Jeong DH, Lee DS, Kim JR, Park SG, Kang MS, Kim YN, Lee KB, Sung MS, Kim KT (2013) Tetraarsenic oxide and cisplatin induce apoptotic synergism in cervical cancer. Oncol Rep 29(4):1540–1546. doi: 10.3892/or.2013.2243 [DOI] [PubMed] [Google Scholar]
  • 54.Kotowski U, Heiduschka G, Brunner M, Erovic BM, Martinek H, Thurnher D (2012) Arsenic trioxide enhances the cytotoxic effect of cisplatin in head and neck squamous cell carcinoma cell lines. Oncol Lett 3(6):1326–1330. doi: 10.3892/ol.2012.643 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Zhang N, Wu ZM, McGowan E, Shi J, Hong ZB, Ding CW, Xia P, Di W (2009) Arsenic trioxide and cisplatin synergism increase cytotoxicity in human ovarian cancer cells: therapeutic potential for ovarian cancer. Cancer Sci 100(12):2459–2464. doi: 10.1111/j.1349-7006.2009.01340.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Li H, Zhu X, Zhang Y, Xiang J, Chen H (2009) Arsenic trioxide exerts synergistic effects with cisplatin on non-small cell lung cancer cells via apoptosis induction. J ExpClin Cancer Res 28:110. doi: 10.1186/1756-9966-28-110 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Muenyi CS, Pinhas AR, Fan TW, Brock GN, Helm CW, States JC (2012) Sodium arsenite +/− hyperthermia sensitizes p53-expressing human ovarian cancer cells to cisplatin by modulating platinum-DNA damage responses. Toxicol Sci 127(1):139–149. doi: 10.1093/toxsci/kfs085 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Muenyi CS, States VA, Masters JH, Fan TW, Helm CW, States JC (2011) Sodium arsenite and hyperthermia modulate cisplatin-DNA damage responses and enhance platinum accumulation in murine metastatic ovarian cancer xenograft after hyperthermic intraperitoneal chemotherapy (HIPEC). J Ovarian Res 4:9. doi: 10.1186/1757-2215-4-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Muenyi CS, Trivedi AP, Helm CW, States JC (2014) Cisplatin plus sodium arsenite and hyperthermia induces pseudo-G1 associated apoptotic cell death in ovarian cancer cells. Toxicol Sci 139(1): 74–82. doi: 10.1093/toxsci/kfu029 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Stevens JB, Abdallah BY, Liu G, Ye CJ, Horne SD, Wang G, Savasan S, Shekhar M, Krawetz SA, Huttemann M, Tainsky MA, Wu GS, Xie Y, Zhang K, Heng HH (2011) Diverse system stresses: common mechanisms of chromosome fragmentation. Cell Death Dis 2:e178. doi: 10.1038/cddis.2011.60 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Stevens JB, Abdallah BY, Regan SM, Liu G, Bremer SW, Ye CJ, Heng HH (2010) Comparison of mitotic cell death by chromosome fragmentation to premature chromosome condensation. Mol Cytogenet 3:20. doi: 10.1186/1755-8166-3-20 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Stevens JB, Liu G, Bremer SW, Ye KJ, Xu W, Xu J, Sun Y, Wu GS, Savasan S, Krawetz SA, Ye CJ, Heng HH (2007) Mitotic cell death by chromosome fragmentation. Cancer Res 67(16):7686–7694. doi: 10.1158/0008-5472.CAN-07-0472 [DOI] [PubMed] [Google Scholar]
  • 63.Liu G, Stevens JB, Horne SD, Abdallah BY, Ye KJ, Bremer SW, Ye CJ, Chen DJ, Heng HH (2014) Genome chaos: survival strategy during crisis. Cell Cycle 13(4):528–537. doi: 10.4161/cc.27378 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Peters JM (2002) The anaphase-promoting complex: proteolysis in mitosis and beyond. Mol Cell 9(5):931–943 [DOI] [PubMed] [Google Scholar]
  • 65.Uhlmann F (2001) Secured cutting: controlling separase at the metaphase to anaphase transition. EMBO Rep 2(6):487–492. doi: 10.1093/embo-reports/kve113 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Li H, Yin C, Zhang B, Sun Y, Shi L, Liu N, Liang S, Lu S, Liu Y, Zhang J, Li F, Li W, Liu F, Sun L, Qi Y (2013) PTTG1 promotes migration and invasion of human non-small cell lung cancer cells and is modulated by miR-186. Carcinogenesis 34(9):2145–2155. doi: 10.1093/carcin/bgt158 [DOI] [PubMed] [Google Scholar]

RESOURCES