Aopwiki

SNAPSHOT

Created at: 2018-02-23 15:17

AOP ID and Title:


AOP 212: Histone deacetylase inhibition leading to testicular toxicity
Short Title: Histone deacetylase inhibition leading to testicular toxicity

Graphical Representation


Authors


Shihori Tanabe, Akihiko Hirose, Takashi Yamada

Division of Risk Assessment, Biological Safety Research Center, National Institute of Health Sciences


Status

Author status OECD status OECD project SAAOP status
Under development: Not open for comment. Do not cite Under Development 1.52 Included in OECD Work Plan

Abstract


Testicular toxicity is of interest for human health risk assessment especially in terms of reproductive and developmental toxicity, however, the testicular toxicity has not fully elucidated. Histone deacetylase (HDAC) inhibitors (HDIs) are approved as anti-cancer drugs since HDIs have apoptotic effect in cancer cells. HDIs includes the short chain fatty acids (e.g., butyrate, valproate, methoxyacetic acid), hydroxamic acids (e.g., suberoylanilide hydroxamic acid or SAHA, Trichostatin A or TSA), cyclic tetrapeptides (e.g., FK-228), benzamides (e.g., N-acetyldinaline and MS-275) and epoxides (depeudecin, trapoxin A), of which MAA especially focused on have the testicular toxicity such as testis atrophy in vivo. The intracellular mechanisms of induction of the spermatocyte apoptosis by HDIs are suggested as HDAC inhibition as molecular initiating event (MIE), histone acetylation increase, p21 expression increase, and cell cycle disorder as key events (KEs). Adverse outcome includes spermatocyte depletion and testis atrophy and weight loss. We propose new adverse outcome pathway (AOP) for histone deacetylase inhibition leading to testicular toxicity.  



Summary of the AOP

Events

Molecular Initiating Events (MIE), Key Events (KE), Adverse Outcomes (AO)

Sequence Type Event ID Title Short name
1 MIE 1502 Histone deacetylase inhibition Histone deacetylase inhibition
2 KE 1503 Histone acetylation, increase Histone acetylation, increase
3 KE 1504 p21 expression, increase p21 expression, increase
4 KE 1505 cell cycle disorder cell cycle disorder
5 KE 1262 Apoptosis Apoptosis
6 AO 1506 spermatocyte depletion, testis atrophy/weight loss (testicular toxicity) testicular toxicity

Key Event Relationships

Upstream Event Relationship Type Downstream Event Evidence Quantitative Understanding
Histone deacetylase inhibition adjacent Histone acetylation, increase High High
Histone acetylation, increase adjacent p21 expression, increase Moderate Moderate
p21 expression, increase adjacent cell cycle disorder High Moderate
cell cycle disorder adjacent Apoptosis High Moderate
Apoptosis adjacent spermatocyte depletion, testis atrophy/weight loss (testicular toxicity) High Moderate
Histone deacetylase inhibition non-adjacent p21 expression, increase High High
Histone deacetylase inhibition non-adjacent cell cycle disorder High High
Histone deacetylase inhibition non-adjacent Apoptosis High High
Histone deacetylase inhibition non-adjacent spermatocyte depletion, testis atrophy/weight loss (testicular toxicity) High High

Stressors


Name Evidence
Methoxyacetic acid High
Butyrate High
Trichostatin A High

Overall Assessment of the AOP

Assessment of the Weight-of-Evidence supporting the AOP

 

Concordance of dose-response relationships

This is a quantitative description on dose-response relationships from MIE to AOP. But some KE relationships individually are not fully supported with dose-response relationships, while there is empirical evidence to support that a change in KEup leads to an appropriate change in the respective KEdown.

 

Temporal concordance among the key events and adverse outcome

Temporal concordance between MIE and AOP has been described with in vivo experimental data. Empirical evidences show temporal concordance between MIE and the individual KEs, however, the temporal concordance among the individual KEs and AO is not fully elucidated.

 

Strength, consistency, and specificity of association of adverse outcome and initiating event

The scientific evidence on the linkage between MIE and AO has been described.

 

Biological plausibility, coherence, and consistency of the experimental evidence

The available data supporting the AOP are logic, coherent and consistent with established biological knowledge, whereas there are possibilities for alternative pathways.

 

Alternative mechanism(s) that logically present themselves and the extent to which they may distract from the postulated AOP

 

There are some other important apoptotic pathways that are involved in cell death, as well as other important spermatocyte signaling or mechanism influences testicular toxicity.

 

p53 pathway

The study in which in vivo administration of trichostatin A (TSA), a HDI, in mice resulted in male meiosis impairment showed the involvement of p53-noxa-caspase-3 apoptotic pathway in TSA-induced spermatocyte apoptosis (Fenic). Other study showed that MAA induced up-regulation of p21 expression is mediated through histone hyperacetylation and independent of p53/p63/p73 (Parajuli).

 

      NF-kappaB pathway

The present AOP focuses on p21 pathway leading to apoptosis, however, the alternative pathway such as NF-kB signaling pathways may be involved in apoptosis of spermatocytes (Wang).

 

Communication with Sertoli cells

The present AOP focuses on testicular atrophy by HDAC inhibition-induced apoptosis in spermatocytes, however, the signaling in Setoli cells may be involved in testicular atrophy. Sertoli cell secretes GDNF, FGF2, CXCL12 or Ccl9 molecules, which results in the activation of RET, FGFR, CXCR4 or CCR1 signaling in spermatogonial stem cells, respectively (Chen SR and Liu Y).

 

Decrease in deoxynucleotide pool by MAA

MAA induces decrease in deoxynucleotide pool, resulting apoptosis, which may be an alternative pathway other than p21-mediated pathway (Yamazoe). Inhibition of 5,10-CH2-THF production by MAA may decreases deoxynucleotide pool in spermatocytes (Yamazoe).

 

Assessment of the quantitative understanding of the AOP

The quantitative understanding of the AOP in terms of indirect relations between HDAC inhibition and testicular atrophy was examined in in vivo experiments (Foster, Miller).


Domain of Applicability

Life Stage Applicability
Life Stage Evidence
Adult, reproductively mature High
Taxonomic Applicability
Term Scientific Term Evidence Links
human Homo sapiens Moderate NCBI
mouse Mus musculus Moderate NCBI
rat Rattus norvegicus High NCBI
Sex Applicability
Sex Evidence
Male High

Life stage applicability

Life stage

Evidence

Adult, reproductively mature

Strong

 

Taxonomic applicability

Name

Scientific name

Evidence

human

Homo sapiens

Moderate

mouse

Mus musculus

Moderate

rat

Rattus norvegicus

Strong

 

Sex applicability

Sex

Evidence

Male

Strong

Essentiality of the Key Events

Molecular Initiating Event

Support for essentiality

Histone deacetylase inhibition

Strong

 

Key Event

Support for essentiality

Histone acetylation, increase

Strong

p21 expression, increase

Moderate

Cell cycle disorder

Moderate

Apoptosis

Moderate

 

Weight of Evidence Summary

Event

Description

Triggers

Weight of Evidence

Histone deacetylase inhibition

Leads to

Histone acetylation, increase

Strong

Histone acetylation, increase

Leads to

p21 expression, increase

Strong

p21 expression, increase

Leads to

Cell cycle disorder

Strong

Cell cycle disorder

Leads to

Apoptosis

Strong

Apoptosis

Leads to

Spermatocyte depletion, Testis atrophy/weight loss (Testicular toxicity)

Moderate

Histone deacetylase inhibition

Indirectly Leads to

p21 expression, increase

Strong

Histone deacetylase inhibition

Indirectly Leads to

Cell cycle disorder

Strong

Histone deacetylase inhibition

Indirectly Leads to

Cell death, apoptosis

Strong

Histone deacetylase inhibition

Indirectly Leads to

Spermatocyte depletion, Testis atrophy/weight loss (Testicular toxicity)

Strong

Quantitative Consideration

Event

Description

Triggers

Quantitative understanding

Histone deacetylase inhibition

Leads to

Histone acetylation, increase

Strong

Histone acetylation, increase

Leads to

p21 expression, increase

Moderate

p21 expression, increase

Leads to

Cell cycle disorder

Moderate

Cell cycle disorder

Leads to

Apoptosis

Moderate

Apoptosis

Leads to

Spermatocyte depletion, Testis atrophy/weight loss (Testicular toxicity)

Moderate

Histone deacetylase inhibition

Indirectly Leads to

p21 expression, increase

Strong

Histone deacetylase inhibition

Indirectly Leads to

Cell cycle disorder

Strong

Histone deacetylase inhibition

Indirectly Leads to

Cell death, apoptosis

Strong

Histone deacetylase inhibition

Indirectly Leads to

Spermatocyte depletion, Testis atrophy/weight loss (Testicular toxicity)

Strong

Considerations for Potential Applications of the AOP (optional)


The present AOP can be used in risk assessment of HDAC inhibitors for the anti-cancer drugs in terms of testicular toxicity. HDAC inhibitors nowadays have been utilized as therapeutics for cancer or neurology disease, and the adverse effects of HDAC inhibitors should be evaluated. This AOP elucidating the pathway from HDAC inhibition through testicular toxicity may provides important insights for potential toxicity of HDAC inhibitors. It also provides a basis for the HDAC inhibition-induced epigenetic alteration and cell death.

References


Fenic I et al. (2008) In vivo application of histone deacetylase inhibitor trichostatin-A impairs murine male meiosis. J Andro 29: 172-185

Parajuli KR et al. (2014) Methoxyacetic acid suppresses prostate cancer cell growth by inducing growth arrest and apoptosis. Am J Clin Exp Urol 2:300-312

Wang C et al. (2017) CD147 regulates extrinsic apoptosis in spermatocytes by modulating NFkB signaling pathways. Oncotarget 8: 3132-3143

Chen S and Liu Y. (2015) Regulation of spermatogonial stem cell self-renewal and spermatocyte meiosis by Sertoli cell signaling. Reproduction 149: R159-R167

Yamazoe Y. et al. (2015) Embryo- and testicular-toxicities of methoxyacetate and the related: a review on possible roles of one-carbon transfer and histone modification. Food Safety 3:92-107

Foster PM et al. (1983) Testicular toxicity of ethylene glycol monomethyl and monoethyl ethers in the rat. Toxicol Appl Pharmacol 69:385-39

Miller RR et al. (1982) Toxicity of methoxyacetic acid in rats. Fundam Appl Toxicol 2: 158-160

Wade MG et al. (2008) Methoxyacetic acid-induced spermatocyte death is associated with histone hyperacetylation in rats. Biol Reprod 78:822-831

Richon VM et al. (2000) Histone deacetylase inhibitor selectively induces p21WAF1 expression and gene-associated histone acetylation. Proc Natl Acad Sci 97:10014-10019       

Struhl K. (1998) Histone acetylation and transcriptional regulatory mechanisms. Gene Dev 12:599-606                        

O’Reilly MA et al (2001) The cyclin-dependent kinase inhibitor p21 protects the lung from oxidative stress. Am J Respir Cell Mol Biol 24: 703-710          

Wu R et al. (2016) microRNA-497 induces apoptosis and suppressed proliferation via the Bcl-2/Bax-caspase9-caspase 3 pathway and cyclin D2 protein in HUVECs. PLoS One 11: e0167052    

Li L et al. (2012) Downregulation of microRNAs miR-1, -206 and -29 stabilizes PAX3 and CCND2 expression in rhabdomyosarcoma. Lab Invest 92: 571-583 

Mermelshtein A et al. (2005) Expression of F-type cyclins in colon cancer and in cell lines from colon carcinomas. Br J Cancer 93: 33          

Dong Q et al. (2010) microRNA let-7a inhibits proliferation of human prostate cancer cells in vitro and in vivo by targeting E2F2 and CCND2. PLoS One 5: e10147

Kerr JFR et al. (1972) Apoptosis: a basic biological phenomenon with wide-ranging implications in tissue kinetics. Br J Cancer 26: 239-257                                              

Lagger G et al. (2002) Essential function of histone deacetylase 1 in proliferation control and CDK inhibitor repression. EMBO J 21:2672-2681                                      

Schroeder FA et al. (2013) A selective HDAC 1/2 inhibitor modulates chromatin and gene expression in brain and alters mouse behavior in two mood-related tests. PLoS One 8:e71323

Choudhary C et al. (2009) Lysine acetylation targets protein complexes and co-regulates major cellular functions. Science 325:834-840

Henderson SE et al. (2016) Suppression of tumor growth and muscle wasting in a transgenic mouse model of pancreatic cancer by the novel histone deacetylase inhibitor AR-42. Neoplasia 18:765-774

Yoshida M et al. (1990) Potent and specific inhibition of mammalian histone deacetylase both in vivo and in vitro trichostatin A. J Biol Chem 265:17174-17179

Gnad F et al. (2011) PHOSIDA 2011: the posttranslational modification database. Nucl Acids Res 39:D253-D260

Allfrey V et al (1964) Acetylation and methylation of histones and their possible role in the regulation of RNA synthesis. Proc Natl Acad Sci 51: 786-794

Pogo B et al (1966) RNA synthesis and histone acetylation during the course of gene activation in lymphocytes. Proc Natl Acad Sci 55: 805-812

Richon VM et al. (2000) Histone deacetylase inhibitor selectively induces p21WAF1 expression and gene-associated histone acetylation. Proc Natl Acad Sci 97:10014-10019

Wu JT et al. (2001) Transient vs prolonged histone hyper acetylation: effects on colon cancer cell growth, differentiation, and apoptosis. Am J Physiol Gastrointest Liver Physiol 280:G482-G490

Gartel AL and Tyner AL (2002) The role of the cyclin-dependent kinase inhibitor p21 in apoptosis. Mol Cancer Ther 1: 639-649

Chen J et al (1996) Cyclin-binding motifs are essential for the function of p21CIP1. Mol Cell Biol 16: 4673-4682                       

Li L et al. (2012) Downregulation of microRNAs miR-1, -206 and -29 stabilizes PAX3 and CCND2 expression in rhabdomyosarcoma. Lab Invest 92: 571-583

Dong Q et al. (2010) microRNA let-7a inhibits proliferation of human prostate cancer cells in vitro and in vivo by targeting E2F2 and CCND2. PLoS One 5: e10147

Wu R et al. (2016) microRNA-497 induces apoptosis and suppressed proliferation via the Bcl-2/Bax-caspase9-caspase 3 pathway and cyclin D2 protein in HUVECs. PLoS One 11: e0167052    

Niu Z et al. (2011) microRNA-21 regulates the self-renewal of mouse spermatogonial stem cells. Proc Natl Acad Sci 108: 12740-12745

de Rooij DG et al. (2001) Proliferation and differentiation of spermatogonial stem cells. Reproduction 121: 347-354

de Rooij DG. (1998) Stem cells in the testis. Int J Exp Path 79: 67-80


Appendix 1

List of MIEs in this AOP

Event: 1502: Histone deacetylase inhibition

Short Name: Histone deacetylase inhibition

AOPs Including This Key Event

AOP ID and Name Event Type
Aop:212 - Histone deacetylase inhibition leading to testicular toxicity MolecularInitiatingEvent

Stressors

Name
Methoxyacetic acid
Butyrate
Trichostatin A

Biological Context

Level of Biological Organization
Molecular

Evidence for Perturbation by Stressor


Overview for Molecular Initiating Event

HDIs are classified according to chemical nature and mode of mechanism: the short chain fatty acids (e.g., butyrate, valproate), hydroxamic acids (e.g., suberoylanilide hydroxamic acid or SAHA, Trichostatin A or TSA), cyclic tetrapeptides (e.g., FK-228), benzamides (e.g., N-acetyldinaline and MS-275) and epoxides (depeudecin, trapoxin A) [Richon VM et al 2004, Ropero S and Esteller M, Villar-Garea et al]. There is a report showing that TSA and butyrate competitively inhibits HDAC activity [Sekhavat A]. HDIs inhibit preferentially HDACs with some selectiveness [Hu E et al]. TSA inhibits HDAC1, HDAC3 and HDAC8, whereas MS-27-275 has inhibitory effect for HDAC1 and HDAC3 (IC50 value of ~0.2 mM and ~8 mM, respectively), but no effect for HDAC8 (IC50 value >10 mM) [Hu E et al]. TSA inhibits HDAC1, 2, 3 of class I HDACs. HDAC 1, 4, 6 are related to tumor size [Damaskos]. MAA (2 or 5 mM) inhibited HDAC activity in dose-response manner in rat testis cytosolic and nuclear extracts [Wade MG 2008].



Domain of Applicability


Taxonomic Applicability
Term Scientific Term Evidence Links
rat Rattus norvegicus High NCBI
human Homo sapiens High NCBI
mouse Mus musculus High NCBI

Name

Scientific Name Tissue/Organ or Cell type Evidence Reference
Rat Rattus norvegicus Brown Norway rat (output: rate of resorption of subretinal blebs) (Desjardins) / Sprague-Dawley rat  (Wade) / pc-12 cells (Steffan JS) Strong

Desjardins D et al.

Wade MG et al 2008

Steffan JS et al.
Human Homo sapiens  Human (Ansari) / NSCLC cells (output: cell growth) (Miyanaga) / HeLa cell (Jansen MS, Di Renzo) / ARPE19 cells (Desjardins) Strong

Ansari J et al., Miyanaga A et al.

Jansen MS et al. 2004

Di Renzo et al.

Desjardins D et al.
Mouse Mus musculus Splenocytes (Mishra) / brain, spleen (Hockly) / spleen (Jansen MS) / embyo (Di Renzo) Strong Mishra N et al. Hockly E et al. Jansen MS et al. 2004 Di Renzo et al.

 


Key Event Description

Site of action: The site of action for the molecular initiating event is the spermatocytes in testis.

The nucleosome consists of core histones having classes of H2A, H2B, H3 and H4) [Damaskos]. DNA strand (about 200 bp) wound around the core histones, where histone deacetylase (HDAC) effects on the lysine residue of the histone to hydrolyze the acetyl residue [Damaskos]. Histone deacetylase inhibitor (HDI) inhibits HDAC and acetylate the histones and release the DNA strand to induce the binding of transcription factors [Taunton et al]. HDIs have potentials as anti-cancer pharmaceuticals since HDIs induce the transcriptional restoration of epigenetically silenced tumor suppressor genes by regulating acetylation of histones and non-histone proteins [Lee et al] [Minucci S].

It is known that 18 HDAC isoforms are classified into four classes: class I HDACs (isoforms 1, 2, 3, 8), class II isoforms (4, 5, 6, 7, 9, 10) and class III HDACs (the sirtuins) and HDAC11 [Weichert, Barneda-Zahonero]. HDACs 1, 2 and 3 are ubiquitously expressed, whereas HDAC8 is predominantly expressed in cells with smooth muscle/myoepithelial differentiation [Weichert]. HDAC6 is not observed to express in lymphocytes, stromal cells and vascular endothelial cells [Weichert]. Class III HDACs sirtuins are widely expressed and localized in different cellular compartments [Barneda-Zahonero]. SirT1 is highly expressed in testis, thymus and multiple types of germ cells [Bell]. HDAC11 expression is enriched in kidney, brain, testis, heart and skeletal muscle [Barneda-Zahonero].


How it is Measured or Detected

The measurement of HDAC inhibition monitors the decrease in histone acetylation. The measurement methods include the immunological detection of histone acetylation with anti-acetylated histone antibodies [Richon VM et al 2004]. The histones are isolated from pellets of cells treated with HDIs, followed by acid-urea-triton gel electrophoresis, western blotting, and immunohistochemistry [Richon VM et al 2004]. Epigenetic modifications including the histone acetylation are measured using chromatin immunoprecipitation-microarray hybridization (ChIP-chip) [ENCODE Project Consortium, Ren B et al]. ChIP detects physical interaction between transcription factors or cofactors and the chromosome [Johnson DS et al].


References

Damaskos C. et al. (2017) Histone deacetylase inhibitors: an attractive therapeutic strategy against breast cancer. Anticancer Research 37: 35-46.

Taunton J. et al. (1996) A mammalian histone deacetylase related to the Yeast transcriptional regulator Rpd3p. Science 272:408-411.

Lee SC. et al. (2016) Essential role of insulin-like growth factor 2 in resistance to histone deacetylase inhibitor. Oncogene 35:5515-5526.

Minucci S, Pelicci PG. (2006) Histone deacetylase inhibitors and the promise of epigenetic (and more) treatments for cancer. Nat Rev Cancer. Jan;6(1):38-51.

Weichert W. (2009) HDAC expression and clinical prognosis in human malignancies. Cancer Letters 280:168-176

Barneda-Zahonero B and Parra M (2012) Histone deacetylases and cancer. Mol Oncol 6:579-589

Bell EL et al. (2014) SirT1 is required in the male germ cell for differentiation and fecundity in mice. Development 141:3495-3504

Richon VM et al. (2004) Histone deacetylase inhibitors: assays to assess effectiveness in vitro and in vivo. Methods Enzymol. 376:199-205

The ENCODE Project Consortium. (2004) The ENCODE (ENCyclopedia Of DNA Elements) Project. Science 306:636-640

Ren B and Dynlacht D. (2004) Use of chromatin immunoprecipitation assays in genome-wide location analysis of mammalian transcription factors. Methods Enzymol. 376:304-315

Johnson DS et al. (2007) Genome-wide mapping of in vivo protein-DNA interactions. Science 316:1497-1502

Desjardins D et al. (2016) Histone deacetylase inhibition restores retinal pigment epithelium function in hyperglycemia. PLoS ONE 11: e0162596

Wade MG et al. (2008) Methoxyacetic acid-induced spermatocyte death is associated with histone hyperacetylation in rats. Biol Reprod 78:822-831

Steffan JS et al. (2001) Histone deacetylase inhibitors arrest polyglutamine-dependent neurodegeneration in Drosophila. Nature 413:739-743

Ansari J et al. (2016) Epigenetics in non-small cell lung cancer: from basics to therapeutics. Transl Lung Cancer Res 5:155-171

Miyanaga A et al. (2008) Antitumor activity of histone deacetylase inhibitors in non-small cell lung cancer cells: development of a molecular predictive model. Mol Cancer Ther 7:1923-1930

Jansen MS et al. (2014) Short-chain fatty acids enhance nuclear receptor activity through mitogen-activated protein kinase activation and histone deacetylase inhibition Proc Natl Acad Sci USA 101:7199-7204

Di Renzo F et al. (2007) Boric acid inhibits embryonic histone deacetylases: A suggested mechanism to explain boric acid-related teratogenicity. Toxicol and Appl Pharmacol 220:178-185

Mishra N et al. (2003) Histone deacetylase inhibitors modulate renal disease in the MRL-lpr/lpr mouse. J Clin Invest 111: 539-552

Hockly E et al. (2003) Suberoylanilide hydroxamic acid, a histone deacetylase inhibitor, ameliorates motor deficits in a mouse model of Huntington’s disease. Proc Nat Acad Sci 100:2041-2046

Ropero S and Esteller M. (2007) The role of histone deacetylases (HDACs) in human cancer. Mol Oncol 1:19-25

Villae-Garea A and Esteller M. (2004) Histone deacetylase inhibitors: understanding a new wave of anticancer agents. Int J Cancer 112:171-178

Sekhavat A et al. (2007) Competitive inhibition of histone deacetylase activity by trichostatin A and butyrate. Biochemistry and Cell Biology 85:751-758

Hu E et al. (2003) Identification of novel isoform-selective inhibitors within class I histone deacetylases. J Pharmacol Exp Ther 307:720-728

Damaskos C et al. (2016) Histone deacetylase inhibitors: a novel therapeutic weapon against medullary thyroid cancer? Anticancer Res 36:5019-5024


List of Key Events in the AOP

Event: 1503: Histone acetylation, increase

Short Name: Histone acetylation, increase

AOPs Including This Key Event


Biological Context

Level of Biological Organization
Cellular

Domain of Applicability


MAA induces acetylation of histones H3 and H4 in Sprague-Dawley (Rattus norvegicus) [Wade MG]. It is also reporeted that MAA promotes acetylation of H4 in HeLa cells (Homo sapiens) and spleens from C57BL/6 mice (Mus musculus) treated with MAA [Jansen]. VPA induces hyperacetylation of histone H4 in protein extract of mouse embryos (Mus musculus) exposed in utero for 1h to VPA [Di Renzo]. Apicidin, MS-275 and sodium butyrate induce hyperacetylation of histone H4 in homogenates from mouse embryos (Mus musculus) after the treatments [Di Renzo]. MAA acetylates histones H3K9 and H4K12 in limbs of CD1 mice (Mus musculus) [Dayan C].


Key Event Description

Gene transcription is regulated with the balance between acetylation and deacetylation. The acetylation and deacetylation are modulated on the NH3+ groups of lysine amino acid residues in histones. DNA in acetylated histones is more accessible for transcription factors, leading to increase in gene expression. HDAC inhibition promotes the hyperacetylation by inhibiting deacetylation of histones with classes of H2A, H2B, H3 and H4 in nucleosomes. [Wade MG 2008].


How it is Measured or Detected

Histone acetylation is measured by the immunological detection of histone acetylation with anti-acetylated histone antibodies [Richon VM et al 2004]. Histone acetylation on chromatin can be measured using labeling method with sodium [3H] acetate [Gunjan A et al].


References

Wade MG et al. (2008) Methoxyacetic acid-induced spermatocyte death is associated with histone hyperacetylation in rats. Biol Reprod 78:822-831

Richon VM et al. (2004) Histone deacetylase inhibitors: assays to assess effectiveness in vitro and in vivo. Methods Enzymol 376:199-205

Gunjan A et al. (2001) Core histone acetylation is regulated by linker histone stoichiometry in vivo. J Biol Chem 276:3635-3640

Jansen MS et al. (2014) Short-chain fatty acids enhance nuclear receptor activity through mitogen-activated protein kinase activation and histone deacetylase inhibition Proc Natl Acad Sci USA 101:7199-7204

Di Renzo F et al. (2007) Boric acid inhibits embryonic histone deacetylases: A suggested mechanism to explain boric acid-related teratogenicity. Toxicol and Appl Pharmacol 220:178-185

Di Renzo F et al. (2007) Relationship between embryonic histonic hyperacetylation and axial skeletal defects in mouse exposed to the three HDAC inhibitors apicidin, MS-275, and sodium butyrate. Toxicol Sci 98:582-588

Dayan C and Hales BF. (2014) Effects of ethylene glycol monomethyl ether and its metabolite, 2-methoxyacetic acid, on organogenesis stage mouse limbs in vitro. Birth Defects Res (Part B) 101:254-261


Event: 1504: p21 expression, increase

Short Name: p21 expression, increase

AOPs Including This Key Event


Biological Context

Level of Biological Organization
Cellular

Domain of Applicability


FK228 up-regulated p21 level in human esophageal cancer TE2 cells (Homo sapiens) [Hoshino I]. MAA induced p21 up-regulation in human prostate cancer cell lines (Homo sapiens) [Parajuli]. MAA increases p21 expression in human bladder carcinoma cells, T24 (Homo sapiens) [Glaser]. MAA up-regulated p21 expression in limbs of CD1 embryonic mice (Mus musculus) [Dayan C].


Key Event Description

p21 (CDKN1A) binds to and inhibits the activity of cyclin-dependent kinase 2 or cyclin-dependent kinase 4 complexes, and regulates cell cycle progression in G1 phase. p21 is important for cell cycle regulation. Acetylation of p21 promoter and p21 mRNA were correlated in valproic acid and analog treatments (Gurvich).  


How it is Measured or Detected

The p21 mRNA is measured with real-time RT-PCR technique using primers for p21 [Dayan C]. Gene expression of p21 is measured with microarray technique using gene chips after cDNA preparation from total RNA extracted from the samples [Glaser, Hoshino]. Protein level of p21 is measured with Western blot analysis using anti-p21 antibody [Parajuli, Glaser].


References

Dayan C and Hales BF. (2014) Effects of ethylene glycol monomethyl ether and its metabolite, 2-methoxyacetic acid, on organogenesis stage mouse limbs in vitro. Birth Defects Res (Part B) 101:254-261

Glaser KB et al. (2003) Gene expression profiling of multiple histone deacetylase (HDAC) inhibitors: defining a common gene set produced by HDAC inhibition in T24 and MDA carcinoma cell lines. Mol Cancer Ther 2:151-163

Hoshino I et al. (2007) Gene expression profiling induced by histone deacetylase inhibitor, FK228, in human esophageal squamous cancer cells. Oncol Rep 18:585-592

Parajuli KR et al. (2014) Methoxyacetic acid suppresses prostate cancer cell growth by inducing growth arrest and apoptosis. Am J Clin Exp Urol 2:300-313


Event: 1505: cell cycle disorder

Short Name: cell cycle disorder

AOPs Including This Key Event


Biological Context

Level of Biological Organization
Cellular

Domain of Applicability


MAA blocks G1/S transition in human prostate cancer cell cycles (Homo sapiens) [Parajuli]. The change in the amounts of cells in G1 phase and S phase of cell cycle was detected in mouse HDAC1-/- fibroblast lines (Mus musculus) [Zupkovitz].


Key Event Description

In spermatocytes, the mitosis and meiosis are activated and regulated. The dysregulation of cell cycle effects the spermatogenesis such as numbers. The phosphorylation of p21 regulates its function [Moussa, Child]. The up-regulation of p21 level in iron-chelated cancer cells was observed [Moussa]. G1/S transition blockade was observed in MAA-treated prostate cancer cells [Parajuli]. Valproic acid (VPA), a well-known teratogen, inhibits HDACs, which leads to the teratogenic effect [Gurvich].


How it is Measured or Detected

Cell cycle analysis was performed with cells treated with HDAC inhibitor [Parajuli]. The percentage of cells at G1, G0, S, and G2/M phases was determined by flow cytometry analysis using DNA content frequency histogram deconvolution software [Parajuli, Li Q]. Cell cycle distribution in HDAC1-/- fibroblast lines was also analyzed by fluorescence-activated cell sorter (FACS) analysis with a Partec PAS-II sorter [Zupkovitz]. The four cell cycle phases in living cells can be measured with four-color fluorescent proteins using live cell imaging [Bajar].


References

Moussa RS et al. (2015) Differential targeting of the cyclin-dependent kinase inhibitor, p21CIP/WAF1, by chelators with anti-proliferative activity in a range of tumor cell-types. Oncotarget 6:29694-29711

Child ES and Mann DJ. (2006) The intricacies of p21 phosphorylation. Cell Cycle 5:1313-1319

Parajuli KR et al. (2014) Methoxyacetic acid suppresses prostate cancer cell growth by inducing growth arrest and apoptosis. Am J Clin Exp Urol 2:300-313

Gurvich N et al. (2004) Histone deacetylase is a target of valproic acid-mediated cellular differentiation. Cancer Research 64:1079-1086

Li Q, Lambrechts MJ, Zhang Q, Liu S, Ge D, Yin R, Xi M and You Z. Glyphosate and AMPA inhibit cancer cell growth through inhibiting intracellular glycine synthesis. Drug Des Devel Ther 2013; 7: 635-643.

Zupkovitz G et al. (2010) The cyclin-dependent kinase inhibitor p21 is a crucial target for histone deacetylase 1 as a regulator of cellular proliferation. Mol Cell Biol 30:1171-1181

Bajar BT et al. (2016) Fluorescent indicators for simultaneous reporting of all four cell cycle phases. Nat Methods 13: 993-996


Event: 1262: Apoptosis

Short Name: Apoptosis

Key Event Component

Process Object Action
apoptotic process increased

Biological Context

Level of Biological Organization
Tissue

Domain of Applicability


The apoptosis and proliferation inhibition induced by MAA was measured in human prostate cancer cell lines (Homo sapiens) [Parajuli]. The cell viability inhibition induced by SAHA or TSA was observed in NHDFs (Homo sapiens) [Glaser]. The proliferation of the HDAC-/- ES cells was inhibited compared to HDAC+/+ ES cells (Homo sapiens) [Zupkovitz].


Key Event Description

Apoptosis is characterized by DNA ladder and chromatin condensation. Several stimuli such as hypoxia, nucleotides deprivation, chemotherapeutical drugs, DNA damage, and mitotic spindle damage induce p53 activation, leading to p21 activation and cell cycle arrest [Pucci]. MAA (5 mM or 20 mM) decreased BIRC2 protein expression and activated caspases 7 and 3 [Parajuli]. MAA induces apoptotic nucleosome increase and cleaved PARP protein expression [Parajuli]. MAA decreased cell viability of human prostate cancer cell lines [Parajuli]. The SAHA or TSA treatment on neonatal human dermal fibroblasts (NHDFs) for 24 or 72 hrs inhibited proliferation/viability of the cells [Glaser]. The impaired proliferation was observed in HDAC-/- ES cells, which was rescued with the reintroduction of HDAC1 [Zupkovitz].


How it is Measured or Detected

The apoptosis is detected with the expression alteration of procaspases 7 and 3 by Western blotting using antibodies [Parajuli]. The apoptosis is measured with down-regulation of anti-apoptotic gene baculoviral inhibitor of apoptosis protein repeat containing 2 (BIRC2, or cIAP1) [Parajuli]. Apoptotic nucleosomes were detected using Cell Death Detection ELISA kit, which were calculated as absorbance subtraction at 405 nm and 490 [Parajuli]. Cell viability was measured with live cell number changes using the CellTiter-Glo Luminescent Cell Viability Assay [Parajuli]. Cleavage of PARP was detected with Western blotting [Parajuli]. The proliferation/viability of NHDFs was measured with Alamar-Blue [modified 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide] [Glaser]. Proliferation of the HDAC-/- ES cells was determined with crystal violet and measurement of absorbance at 595 nm [Zupkovitz].


References

Pucci B et al. (2000) Cell cycle and apoptosis. Neoplasia 2:291-299

Parajuli KR et al. (2014) Methoxyacetic acid suppresses prostate cancer cell growth by inducing growth arrest and apoptosis. Am J Clin Exp Urol 2:300-313

Glaser KB et al. (2003) Gene expression profiling of multiple histone deacetylase (HDAC) inhibitors: defining a common gene set produced by HDAC inhibition in T24 and MDA carcinoma cell lines. Mol Cancer Ther 2:151-163

Zupkovitz G et al. (2010) The cyclin-dependent kinase inhibitor p21 is a crucial target for histone deacetylase 1 as a regulator of cellular proliferation. Mol Cell Biol 30:1171-1181


List of Adverse Outcomes in this AOP

AOPs Including This Key Event


Biological Context

Level of Biological Organization
Organ

Domain of Applicability


It has been reported that mice lacking both Ink4c and Ink4d produced few mature sperm, and the residual spermatozoa had reduced motility and decreased viability (Mus musculus) [Zindy]. The sperm counts in the cauda epidydimis of rats exposed to butylparaben were significantly decreased (Rattus norvegicus) [Oishi S2001]. EGME or MAA treatment induced the testicular damage in rat (Rattus norvegicus) [Foster PMD1983]. EGME were shown to deplete the spermatocytes in CD-1 mice (Mus musculus) and CD rats (Rattus norvegicus), principally pachytene cells, but with other stages affected with increasing dose [Anderson D]. The testicular lesions induced by 2-methoxyethanol were observed in rats (Rattus norvegicus) and guinea pigs (Cavia porcellus), which are different in onset, characteristics and severity [Ku WW]. EGME has effects in disruption of spermatogenesis in rabbits (Oryctolagus cuniculus) [Foote]. Dimethoxyhexane (DMH) induces testicular toxicity such as spermatocyte death in seminiferous tubule stages I-IV and stages XII-XIV and MAA increase in urine in Sprague-Dawley rats (Rattus norvegicus) MAA treatment induces spermatocyte death in Sprague-Dawley rats (Rattus norvegicus) [Wade 2008]. Treatment of TSA resulted in a dose-dependent decrease in relative testis weight due to impaired spermatogenesis in mice, and impaired meiosis [Fenic 2004, 2008].


Key Event Description

Spermatocytes are differentiated from spermatogonial stem cells via random proliferation, differentiation and synchronized mitoses with several stages [Rooij]. Spermatogenesis is controlled by the cyclin D-dependent kinase inhibitors p18Ink4c and p19Ink4d [Zindy]. It is hypothesized that the testicular effects of 1,6-dimethoxyhexane (DMH) are caused by its metabolism to MAA [Wade2006, Poon2004]. MAA produces testicular and thymic atrophy [Miller1982, Moss1985]. The spermatogenic stages in which the toxicity of MAA is induced are on the patchytene spermatocytes immediately before and during meiotic division, which are Stages XII-XIV of the cycle in the rat and the early pachytene spermatocytes at stages I-IV of the cycle. Dead germ cells can be seen as soon as 12 hours after the treatment of MAA [Casarett & Doull’s].


How it is Measured or Detected

The sperm-containing fluid was squeezed out of the cauda, and suspended in medium containing HEPEs buffer and bovine serum albumin, and incubated at 37C for 20 min. The number of spermatozoa was determined by hematocytometer [Zindy]. Testicular sperm counts and daily sperm production were determined by counting the total number of spermatids per testis, and divided by the testicular weight to give the results in spermatids per gram of testis [Oishi S2001]. The weights of testes of MAA-treated rats were measured to detect the testicular atrophy [Foster PMD1983]. Since zinc concentration has been shown to play an important role in the production of testicular injury by compounds, the effects of EGME and MAA on urinary zinc excretion and testicular zinc content was examined [Foster PMD1983]. In detail, the animals were ip injected with 65Zn-Cl2 in saline 2 days before treatment with either EGM or MAA. After the measurement of baseline values of 65Zn urinary excretion, groups of animals were administered with EGME or an equimolar dose of MAA by oral gavage for 4 days and urine samples were collected daily [Foster PMD1983]. Twenty-four hours after the last dose of EGME or MAA, the animals were killed and the 65Zn content in urine and tissues was measured with a Packard 5230 autogamma spectrometer [Foster PMD1983]. Testis were fixed in Bouin’s fluid for light microscopy or diced and fixed in a 4% formaldehyde plus 1% glutaraldehyde solution in 0.1 M phosphate buffer, pH 7.29 for transmission electron microscopy [Foster PMD1983, McDowell and Trump1976]. Changes in sperm were measured by computer-assisted sperm analysis [Foote RH]. For the detection of apoptosis, the testes were fixed in modified Davidson fix for 48 hrs, postfixed for 24 hrs in 10% neutral buffered formalin, and embedded in paraffin. Germ cell death was visualized in testis sections by TUNEL staining [Wade2008]. The incidence of TUNEL-positive cells was expressed as the number of positive cells per tubule examined for one entire testis section per animal [Wade2008]. For the testis cell analysis, fresh testes were dispersed using a two-stage enzymatic digestion and incubated in BSA containing collagenase and DNase I [Wade2006]. The seminiferous tubules were further digested and cells were fixed in ice-cold 70% ethanol [Wade2006]. Relative proportions of spermatogenic cell populations were assessed in fixed cells using a flow cytometeric method [Wade2006]. The principle of the test is that spermatogenic cells, as they differentiate from normal diploid spermatogonial stem cells through to mature spermatozoa with a highly condensed haploid complement of DNA, progress through various intermediate stages with differing nuclear DNA content and cellular content of mitochondria. With the use of fluorescent probes to quantitatively label DNA (PI) and mitochondria (NAO), the relative numbers of cells in each intermediate population can be resolved to determine relatives at each germ cell stage. Cells were analyzed using a FACS Calibur flow cytometer fitted with an argon ion laser (488 nm line excitation); fluorescence emission of NAO was reflected by a 550 dichroic longpass filter and quantified after passage through a 530/30 nm bandpass filter [Wade2006]. Fluorescence of PI was detected after sequential passage through the 550 dichroic filter and a 670-nm longpass filter [Wade2006]. Relative proportions of cells in each population were calculated with WinList software [Wade2006]. The germ cell types in each population were identified under a fluorescent microscope (Zeiss Axioskop 2, Carl Zeiss, Thornwood, NY) by examination of cells from each population, which were isolated using the cell-sorting capacity of the FACS Calibur flow cytometer [Wade2006]. For the assessment of sperm morphology, eosin-stained sperm collected from the cauda epididymis were smeared onto two glass slides per sample, air-dried, and cover-slipped. At least 200 sperm on each slide were examined for the proportion of sperm with abnormal head (overhooked, blunt hook, banana-shaped, amorphous, or extremely oversized) or tail (twisted, bent, corkscrew, double. multiple) by one individual unaware of animal number or treatment [Wade2006]. For the measurement of the total number of condensed spermatids per testis, a weighed portion of the parenchyma from the left testis, as representative of the whole organ as possible, was homogenized in 20 ml of STA solution (0.9% NaCl, 0.01% Triton X-100, and 0.025% sodium azide) [Wade2006]. For the measurement of the total number of sperm in the cauda epididymis, whole cauda and associated sperm suspension in DPBS were thawed on ice and homogenized [Wade2006]. Homogenates of either tissue were then disrupted with a VibraCell sonicator using a microprobe [Wade2006]. Sperm or homogenization-resistant spermatid nuclei densities were calculated from the average number of nuclei in four fields on a Neubaur hemocytometer and were expressed as total or as per gram of epididymis or testis weight [Wade2006]. For the determination of total LDH and LDH-X in supernatant of the homogenized testis fragment, enzyme activity was measured by monitoring extinction of NAD absorbance at 340 nm in a reaction mixture that contained 4.2 mM of NAD in 10.5 mM Tris KCl (pH 9.0) at 30C [Wade2006].


References

Rooij DG. (2001) Proliferation and differentiation of spermatogonial stem cells. Reproduction 121:347-354

Zindy F et al. (2001) Control of spermatogenesis in mice by the cyclin D-dependent kinase inhibitors p18Ink4c and p19Ink4d. Mol Cell Biol 21:3244-3255

Wade MG et al. (2006) Testicular toxicity of candidate fuel additive 1,6-dimethoxyhexane: comparison with several similar aliphatic ethers. Toxicol Sci 89:304-313

Poon R et al. (2004) Short-term oral toxicity of pentyl ether, 1,4-diethoxybutane, and 1,6-dimethoxyhexane in male rats. Toxicol Sci 77:142-150

Miller R et al. (1982) Toxicity of methoxyacetic acid in rats. Fundam Appl Toxicol 2:158-160

Moss EJ et al. (1985) The role of metabolism in 2-methoxyethanol-induced testicular toxicity. Toxicol Appl Pharmacol 79:480-489

Casarett & Doull’s Toxicology, the Basic Science of Poisons, 7th Edition, Edited by Curtis D. Klaassen, Chapter 20 Toxic responses of the reproductive system

Oishi S. (2001) Effects of butylparaben on the male reproductive system in rats. Toxicol Indust Health 17:31-39

Foster PM et al. (1983) Testicular toxicity of ethylene glycol monomethyl and monoethyl ethers in the rats. Toxicol Appl Pharmacol 69:385-399

McDowell EM and Trump BF. (1976) Histologic fixatives suitable for diagnostic light and electron microscopy. Arch Pathol Lab Med 100:405-414

Foote RH et al. (1995) Ethylene glycol monomethyl ether effects on health and reproduction in male rabbits. Reprod Toxicol 9:527-539

Wade MG et al. (2008) Methoxyacetic acid-induced spermatocyte death is associated with histone hyperacetylation in rats. Biol Reprod 78:822-831

Anderson D et al. (1987) Effect of ethylene glycol monomethyl ether on spermatogenesis, dominant lethality, and F1 abnormalities in the rat and the mouse after treatment of F0 males. Teratog Carcinog Mutagen 7:141-158

Ku WW et al. (1994) Comparison of the testicular effects of 2-methoxyethanol (ME) in rats and guinea pigs. Exp Mol Pathol 61:119-133

Fenic I et al. (2008) In vivo application of histone deacetylase inhibitor trichostatin-A impairs murine male meiosis. J Androl 29: 172-185

Fenic I et al. (2004) In vivo effects of histone-deacetylase inhibitor trichostatin-A on murine spermatogenesiss. J Androl 25: 811-818


Appendix 2

List of Key Event Relationships in the AOP