Research Article
Open access
Published on 9 September 2024
Download pdf
Li,J. (2024). What is the impact of histone modification-based treatment in cardiovascular diseases in modern medicine?. Journal of Food Science, Nutrition and Health,2,40-53.
Export citation

What is the impact of histone modification-based treatment in cardiovascular diseases in modern medicine?

Jingxuan Li *,1,
  • 1 Jinan Foreign Language School

* Author to whom correspondence should be addressed.

https://doi.org/10.54254/3029-0821/2/2024013

Abstract

Cardiovascular diseases (CVDs) are a serious group of diseases worldwide1. Currently, there are a vast number of treatments, including coronary bypass surgery, beta-blockers, diuretics, vitamin C and ACE inhibitors. However, they appear to have unique side effects despite significant therapeutic effects, such as hypokalaemia and cogitative disorders in some patients, especially those with other conditions2. Under these circumstances, effective and targeted drugs against CVDs are in high need of development. Histone modification, as a novel and rapidly developing field, has already attracted many researchers to investigate its role in CVD outbreaks and potential therapeutic effects against CVDs. However, there are currently no certified histone modification-based drugs in CVD therapies. This dissertation will focus on histone modification mechanisms, comparison with traditional CVD drugs, clinical prospects and potential investigation directions as well as current limitations.

Keywords

cardiovascular diseases (CVDs), histone modification-based treatment, modern medicine

[1]. Timmis, A., et al. (2020). European Society of Cardiology: Cardiovascular disease statistics 2019. European Heart Journal, 41(12), 12–85.

[2]. Spn, M., Newman, S., Newman, S. P., & Harrison, M. J. (2002). Coronary-artery bypass surgery and the brain: Persisting concerns. The Lancet Neurology, 1. http://neurology.thelancet.com

[3]. Bhatnagar, A. (2017). Environmental determinants of cardiovascular disease. Circulation Research, 121, 162–180. https://doi.org/10.1161/CIRCRESAHA.117.306458

[4]. Wang, N., et al. (2022). Histone lactylation boosts reparative gene activation post-myocardial infarction. Circulation Research, 131, 893–908.

[5]. Wu, Y., et al. (2023). Phosphoglycerate dehydrogenase activates PKM2 to phosphorylate histone H3T11 and attenuate cellular senescence. Nature Communications, 14.

[6]. Shi, Y., et al. (2022). Epigenetic regulation in cardiovascular disease: Mechanisms and advances in clinical trials. Signal Transduction and Targeted Therapy, 7. https://doi.org/10.1038/s41392-022-01055-2

[7]. Chistiakov, D. A., Orekhov, A. N., & Bobryshev, Y. V. (2017). Treatment of cardiovascular pathology with epigenetically active agents: Focus on natural and synthetic inhibitors of DNA methylation and histone deacetylation. International Journal of Cardiology, 227, 66–82. https://doi.org/10.1016/j.ijcard.2016.11.204

[8]. Zillich, A. J., Garg, J., Basu, S., Bakris, G. L., & Carter, B. L. (2006). Thiazide diuretics, potassium, and the development of diabetes: A quantitative review. Hypertension, 48(2), 219–224.

[9]. Gutiérrez-Repiso, C., et al. (2021). Epigenetic biomarkers of transition from metabolically healthy obesity to metabolically unhealthy obesity phenotype: A prospective study. International Journal of Molecular Sciences, 22.

[10]. Berger, S. L., Kouzarides, T., Shiekhattar, R., & Shilatifard, A. (2009). An operational definition of epigenetics. Genes & Development, 23(7), 781–783.

[11]. Liu, S., et al. (2021). Fat mass and obesity-associated protein regulates RNA methylation associated with depression-like behavior in mice. Nature Communications, 12.

[12]. Zehender, A., et al. (2021). TGFβ promotes fibrosis by MYST1-dependent epigenetic regulation of autophagy. Nature Communications, 12.

[13]. Lawrence, M., Daujat, S., & Schneider, R. (2016). Lateral thinking: How histone modifications regulate gene expression. Trends in Genetics.

[14]. Kim, G. H., Ryan, J. J., & Archer, S. L. (2013). The role of redox signaling in epigenetics and cardiovascular disease. Antioxidants & Redox Signaling, 18(14), 1920–1936.

[15]. Koprinarova, M., Schnekenburger, M., & Diederich, M. (2016). Role of histone acetylation in cell cycle regulation. Current Topics in Medicinal Chemistry, 16(7), 732–744.

[16]. Celano, M., et al. (2018). Targeting post-translational histone modifications for the treatment of non-medullary thyroid cancer. Molecular and Cellular Endocrinology, 469, 38–47. https://doi.org/10.1016/j.mce.2017.05.036

[17]. Yang, Y., Luan, Y., Yuan, R. X., & Luan, Y. (2021). Histone methylation-related therapeutic challenge in cardiovascular diseases. Frontiers in Cardiovascular Medicine, 8. https://doi.org/10.3389/fcvm.2021.710053

[18]. Barros, C. H. N., et al. (2021). Synthesis and self-assembly of curcumin-modified amphiphilic polymeric micelles with antibacterial activity. Journal of Nanobiotechnology, 19.

[19]. Mäntylä, E. (2018). Import and impact: Characterization of parvovirus-nucleus interactions.

[20]. Audia, J. E., & Campbell, R. M. (2016). Histone modifications and cancer. Cold Spring Harbor Perspectives in Biology, 8.

[21]. Tompkins, B. A., & Salerno, T. A. (2021). Commentary: What does the future hold for coronary bypass surgery? Journal of Thoracic and Cardiovascular Surgery, 162, 1120–1121. https://doi.org/10.1016/j.jtcvs.2020.01.029

[22]. Head, S. J., Milojevic, M., Taggart, D. P., & Puskas, J. D. (2017). Current practice of state-of-the-art surgical coronary revascularization. Circulation, 136(12), 1331–1345.

[23]. Head, S. J., & Kappetein, A. P. (2019). Coronary bypass surgery — An ART for dedicated surgeons. New England Journal of Medicine, 380, 489–491.

[24]. Brierley, J. B. (1963). Neuropathological findings in patients dying after open-heart surgery. Thorax, 18.

[25]. Perros, A. J., et al. (2020). Coronary artery bypass grafting is associated with immunoparalysis of monocytes and dendritic cells. Journal of Cellular and Molecular Medicine, 24(9), 4791–4803.

[26]. An, P., et al. (2022). Micronutrient supplementation to reduce cardiovascular risk. Journal of the American College of Cardiology, 80(23), 2269–2285.

[27]. Mosca, L., et al. (1997). Antioxidant nutrient supplementation reduces the susceptibility of low-density lipoprotein to oxidation in patients with coronary artery disease.

[28]. Lykkesfeldt, J., Michels, A. J., & Frei, B. (2014). Vitamin C. Advances in Nutrition, 5(1), 16–18.

[29]. Moser, M. A., & Chun, O. K. (2016). Vitamin C and heart health: A review based on findings from epidemiologic studies. International Journal of Molecular Sciences, 17(8). https://doi.org/10.3390/ijms17081328

[30]. Chen, X., et al. (2023). Copper homeostasis and copper-induced cell death in the pathogenesis of cardiovascular disease and therapeutic strategies. Cell Death & Disease, 14. https://doi.org/10.1038/s41419-023-05639-w

[31]. Oparil, S., et al. (2018). Hypertension. Nature Reviews Disease Primers, 4. https://doi.org/10.1038/nrdp.2018.14

[32]. Fihn, S. D., et al. (2012). 2012 ACCF/AHA/ACP/AATS/PCNA/SCAI/STS guideline for the diagnosis and management of patients with stable ischemic heart disease. Journal of the American College of Cardiology, 60.

[33]. Laurent, S. (2017). Antihypertensive drugs. Pharmacological Research, 124, 116–125. https://doi.org/10.1016/j.phrs.2017.07.026

[34]. Weinberger, M. H. (n.d.). Diuretics and their side effects dilemma in the treatment of hypertension. Hypertension. http://hyper.ahajournals.org/

[35]. Ho, K. M., & Power, B. M. (2010). Benefits and risks of furosemide in acute kidney injury. Anaesthesia, 65(3), 283–293. https://doi.org/10.1111/j.1365-2044.2009.06228.x

[36]. Mas, J.-L., et al. (2017). Patent foramen ovale closure or anticoagulation vs. antiplatelets after stroke. New England Journal of Medicine, 377(11), 1011–1021.

[37]. Xia, S., et al. (2020). Sex differences in primary and secondary prevention of cardiovascular disease in China. Circulation, 141(7), 530–539.

[38]. Kohli, P., et al. (2014). Discharge aspirin dose and clinical outcomes in patients with acute coronary syndromes treated with prasugrel versus clopidogrel: An analysis from the TRITON-TIMI 38 study (trial to assess improvement in therapeutic outcomes by optimizing platelet inhibition with prasugrel-thrombolysis in myocardial infarction 38). Journal of the American College of Cardiology, 63(3), 225–232.

[39]. Johnston, S. C., et al. (2019). Time course for benefit and risk of clopidogrel and aspirin after acute transient ischemic attack and minor ischemic stroke. Circulation, 140(8), 658–664. https://doi.org/10.1161/CIRCULATIONAHA.119.040713

[40]. Khyzha, N., Alizada, A., Wilson, M. D., & Fish, J. E. (2017). Epigenetics of atherosclerosis: Emerging mechanisms and methods. Trends in Molecular Medicine, 23(4), 332–347. https://doi.org/10.1016/j.molmed.2017.02.008

[41]. Xu, S., Pelisek, J., & Jin, Z. G. (2018). Atherosclerosis is an epigenetic disease. Trends in Endocrinology & Metabolism, 29(11), 739–742. https://doi.org/10.1016/j.tem.2018.09.003

[42]. Zhang, D., et al. (2019). Metabolic regulation of gene expression by histone lactylation. Nature, 574(7779), 575–580. https://doi.org/10.1038/s41586-019-1678-1

[43]. Yunna, C., Mengru, H., Lei, W., & Weidong, C. (2020). Macrophage M1/M2 polarization. European Journal of Pharmacology, 877. https://doi.org/10.1016/j.ejphar.2020.173090

[44]. Chen, M. S., Lee, R. T., & Garbern, J. C. (2022). Senescence mechanisms and targets in the heart. Cardiovascular Research, 118(5), 1173–1187. https://doi.org/10.1093/cvr/cvab288

[45]. Man, A. W. C., Li, H., & Xia, N. (2019). The role of Sirtuin1 in regulating endothelial function, arterial remodeling and vascular aging. Frontiers in Physiology, 10. https://doi.org/10.3389/fphys.2019.00918

[46]. Sessa, W. C. (2004). eNOS at a glance. Journal of Cell Science, 117(12), 2427–2429. https://doi.org/10.1242/jcs.01245

[47]. Gan, Y., et al. (2005). Role of histone deacetylation in cell-specific expression of endothelial nitric-oxide synthase. Journal of Biological Chemistry, 280(17), 16467–16475. https://doi.org/10.1074/jbc.M414065200

[48]. Hesham, H. M., Lasheen, D. S., & Abouzid, K. A. M. (2018). Chimeric HDAC inhibitors: Comprehensive review on the HDAC-based strategies developed to combat cancer. Medicinal Research Reviews, 38(6), 2058–2109. https://doi.org/10.1002/med.21488

[49]. Yoon, S., & Eom, G. H. (2016). HDAC and HDAC inhibitor: From cancer to cardiovascular diseases. Chonnam Medical Journal, 52(1), 1–11. https://doi.org/10.4068/cmj.2016.52.1.1

[50]. Ooi, J. Y. Y., et al. (2015). HDAC inhibition attenuates cardiac hypertrophy by acetylation and deacetylation of target genes. Epigenetics, 10(5), 418–430. https://doi.org/10.1080/15592294.2015.1039227

[51]. Han, Y., Nie, J., Wang, D. W., & Ni, L. (2022). Mechanism of histone deacetylases in cardiac hypertrophy and its therapeutic inhibitors. Frontiers in Cardiovascular Medicine, 9. https://doi.org/10.3389/fcvm.2022.887258

[52]. Liu, C. F., & Tang, W. H. W. (2019). Epigenetics in cardiac hypertrophy and heart failure. JACC: Basic to Translational Science, 4(8), 976–993. https://doi.org/10.1016/j.jacbts.2019.09.005

[53]. Kurdi, M., & Booz, G. W. (2011). Three 4-letter words of hypertension-related cardiac hypertrophy: TRPC, mTOR, and HDAC. Journal of Molecular and Cellular Cardiology, 50(6), 964–971. https://doi.org/10.1016/j.yjmcc.2010.11.016

[54]. Zhao, T., et al. (2021). Selective HDAC8 inhibition attenuates isoproterenol-induced cardiac hypertrophy and fibrosis via p38 MAPK pathway. Frontiers in Pharmacology, 12. https://doi.org/10.3389/fphar.2021.673859

[55]. Li, P., Ge, J., & Li, H. (2020). Lysine acetyltransferases and lysine deacetylases as targets for cardiovascular disease. Nature Reviews Cardiology, 17(2), 96–115. https://doi.org/10.1038/s41569-019-0256-5

[56]. Chen, C.-N., et al. (2023). Restoration of Foxp3+ regulatory T cells by HDAC-dependent epigenetic modulation plays a pivotal role in resolving pulmonary arterial hypertension pathology. American Journal of Respiratory and Critical Care Medicine, 208. https://doi.org/10.1164/rccm.202305-0843OC

[57]. Molife, R., et al. (2007). HDAC inhibitors and cardiac safety. Clinical Cancer Research, 13(4), 1068. https://doi.org/10.1158/1078-0432.CCR-06-2283

[58]. Lkhagva, B., et al. (2016). Targeting histone deacetylases: A novel therapeutic strategy for atrial fibrillation. European Journal of Pharmacology, 781, 250–257. https://doi.org/10.1016/j.ejphar.2016.04.027

[59]. Scholz, B., et al. (2019). HDAC (histone deacetylase) inhibitor valproic acid attenuates atrial remodeling and delays the onset of atrial fibrillation in mice. Circulation: Arrhythmia and Electrophysiology, 12. https://doi.org/10.1161/CIRCEP.118.007071

[60]. Lee, D. Y., & Chiu, J. J. (2019). Atherosclerosis and flow: Roles of epigenetic modulation in vascular endothelium. Journal of Biomedical Science, 26. https://doi.org/10.1186/s12929-019-0522-x

[61]. Ho, T. C. S., Chan, A. H. Y., & Ganesan, A. (2020). Thirty years of HDAC inhibitors: 2020 insight and hindsight. Journal of Medicinal Chemistry, 63(21), 12460–12484. https://doi.org/10.1021/acs.jmedchem.0c00487

[62]. Dawson, M. A., & Kouzarides, T. (2012). Cancer epigenetics: From mechanism to therapy. Cell, 150(1), 12–27. https://doi.org/10.1016/j.cell.2012.06.013

[63]. Kumar, A., et al. (2023). Major phytochemicals: Recent advances in health benefits and extraction method. Molecules, 28. https://doi.org/10.3390/molecules28010324

[64]. Petrovski, G., Gurusamy, N., & Das, D. K. (2011). Resveratrol in cardiovascular health and disease. Annals of the New York Academy of Sciences, 1215, 22–33. https://doi.org/10.1111/j.1749-6632.2010.05850.x

[65]. Evans, L. W., Athukorala, M., Martinez-Guryn, K., & Ferguson, B. S. (2020). The role of histone acetylation and the microbiome in phytochemical efficacy for cardiovascular diseases. International Journal of Molecular Sciences, 21(23), 1–18. https://doi.org/10.3390/ijms21239249

[66]. Indira Priyadarsini, K. (2014). The chemistry of curcumin: From extraction to therapeutic agent. Molecules, 19. https://doi.org/10.3390/molecules190912300

[67]. Tian, B., & Liu, J. (2020). Resveratrol: A review of plant sources, synthesis, stability, modification and food application. Journal of the Science of Food and Agriculture, 100(4), 1392–1404. https://doi.org/10.1002/jsfa.10152

[68]. Kumar Singh, A., Bishayee, A., & Pandey, A. K. (2018). Targeting histone deacetylases with natural and synthetic agents: An emerging anticancer strategy. Nutrients, 10(6), 731. https://doi.org/10.3390/nu10060731

[69]. Lu, L., Liu, M., Sun, R. R., Zheng, Y., & Zhang, P. (2015). Myocardial infarction: Symptoms and treatments. Cell Biochemistry and Biophysics, 72(3), 865–867. https://doi.org/10.1007/s12013-015-0533-4

[70]. Taqueti, V. R., & O’Gara, P. T. (2015). Beta-blocker therapy after myocardial infarction: More questions than answers. Journal of the American College of Cardiology, 66(13), 1442–1444. https://doi.org/10.1016/j.jacc.2015.08.002

[71]. Arslan, F. B., Ozturk, K., & Calis, S. (2021). Antibody-mediated drug delivery. International Journal of Pharmaceutics, 596.

[72]. Chang, S. F., et al. (2022). Blood reflux-induced epigenetic factors HDACs and DNMTs are associated with the development of human chronic venous disease. International Journal of Molecular Sciences, 23.

[73]. David Strain, W., & Paldánius, P. M. (2018). Diabetes, cardiovascular disease and the microcirculation. Cardiovascular Diabetology, 17(57).

[74]. Ding, Y., et al. (2022). Application of lipid nanovesicle drug delivery system in cancer immunotherapy. Journal of Nanobiotechnology, 20, 214.

[75]. Drummond, D. C., et al. (2005). Enhanced pharmacodynamic and antitumor properties of a histone deacetylase inhibitor encapsulated in liposomes or ErbB2-targeted immunoliposomes. Clinical Cancer Research, 11, 3392–3401.

[76]. Dyck, G. J. B., Raj, P., Zieroth, S., Dyck, J. R. B., & Ezekowitz, J. A. (2019). The effects of resveratrol in patients with cardiovascular disease and heart failure: A narrative review. International Journal of Molecular Sciences, 20.

[77]. Eaton, D. M., et al. (2022). HDAC inhibition regulates cardiac function by increasing myofilament calcium sensitivity and decreasing diastolic tension. Pharmaceutics, 14.

[78]. Elagizi, A., Kachur, S., Carbone, S., Lavie, C. J., & Blair, S. N. (2020). A review of obesity, physical activity, and cardiovascular disease. Current Obesity Reports, 9, 571–581.

[79]. Evans, L. W., Athukorala, M., Martinez-Guryn, K., & Ferguson, B. S. (2020). The role of histone acetylation and the microbiome in phytochemical efficacy for cardiovascular diseases. International Journal of Molecular Sciences, 21.

[80]. Fu, Z., Li, S., Han, S., Shi, C., & Zhang, Y. (2022). Antibody drug conjugate: The ‘biological missile’ for targeted cancer therapy. Signal Transduction and Targeted Therapy, 7.

[81]. Guasch-Ferré, M., et al. (2020). Glycolysis/gluconeogenesis- and tricarboxylic acid cycle-related metabolites, Mediterranean diet, and type 2 diabetes. American Journal of Clinical Nutrition, 111, 835–844.

[82]. Gunassekaran, G. R., Poongkavithai Vadevoo, S. M., Baek, M. C., & Lee, B. (2021). M1 macrophage exosomes engineered to foster M1 polarization and target the IL-4 receptor inhibit tumor growth by reprogramming tumor-associated macrophages into M1-like macrophages. Biomaterials, 278.

[83]. Hong, J., et al. (2021). F. nucleatum targets lncRNA ENO1-IT1 to promote glycolysis and oncogenesis in colorectal cancer. Gut, 70, 2123–2137.

[84]. Jiang, J., et al. (2021). Lactate modulates cellular metabolism through histone lactylation-mediated gene expression in non-small cell lung cancer. Frontiers in Oncology, 11.

[85]. Li, H., et al. (2021). Artificial exosomes mediated spatiotemporal-resolved and targeted delivery of epigenetic inhibitors. Journal of Nanobiotechnology, 19, 364.

[86]. Liberti, M. V., & Locasale, J. W. (2020). Histone lactylation: A new role for glucose metabolism. Trends in Biochemical Sciences, 45, 179–182.

[87]. Lindemann, H., et al. (2020). Polysaccharide nanoparticles bearing HDAC inhibitor as nontoxic nanocarrier for drug delivery. Macromolecular Bioscience, 20.

[88]. Lindemann, H., et al. (2023). HDACi delivery systems based on cellulose valproate nanoparticles. Methods in Molecular Biology, 2589, 195–205.

[89]. Lovelock, S. L., et al. (2022). The road to fully programmable protein catalysis. Nature, 606, 49–58.

[90]. Maniu, C. V., & Redfield, M. M. (2001). Diastolic dysfunction: Insights into pathophysiology and pharmacotherapy. Expert Opinion on Pharmacotherapy, 2, 997–1008.

[91]. Melissaropoulos, K., et al. (2020). Primary Sjögren’s syndrome and cardiovascular disease. Current Vascular Pharmacology, 18, 447–454.

[92]. Pan, L., et al. (2022). Demethylzeylasteral targets lactate by inhibiting histone lactylation to suppress the tumorigenicity of liver cancer stem cells. Pharmacological Research, 181.

[93]. Pivari, F., et al. (2022). Curcumin supplementation (Meriva®) modulates inflammation, lipid peroxidation and gut microbiota composition in chronic kidney disease. Nutrients, 14.

[94]. Qin, S., et al. (2017). Efficacy and safety of turmeric and curcumin in lowering blood lipid levels in patients with cardiovascular risk factors: A meta-analysis of randomized controlled trials. Nutrition Journal, 16.

[95]. Rashid, M., Brim, H., & Ashktorab, H. (2022). Saffron, its active components, and their association with DNA and histone modification: A narrative review of current knowledge. Nutrients. https://doi.org/10.3390/nu14163317

[96]. Shah, R. R. (2019). Safety and tolerability of histone deacetylase (HDAC) inhibitors in oncology. Drug Safety, 42, 235–245.

[97]. Silveira Rossi, J. L., et al. (2022). Metabolic syndrome and cardiovascular diseases: Going beyond traditional risk factors. Diabetes/Metabolism Research and Reviews, 38.

[98]. Sturgeon, K. M., et al. (2019). A population-based study of cardiovascular disease mortality risk in US cancer patients. European Heart Journal, 40, 3889–3897.

[99]. Tong, X., et al. (2021). Generative models for de novo drug design. Journal of Medicinal Chemistry, 64, 14011–14027.

[100]. Torre, L. A., Siegel, R. L., Ward, E. M., & Jemal, A. (2016). Global cancer incidence and mortality rates and trends: An update. Cancer Epidemiology, Biomarkers & Prevention, 25, 16–27.

[101]. Waddell, I. S., & Orfila, C. (2023). Dietary fiber in the prevention of obesity and obesity-related chronic diseases: From epidemiological evidence to potential molecular mechanisms. Critical Reviews in Food Science and Nutrition, 63.

[102]. Wang, J., et al. (2022). Enterobacterial LPS-inducible LINC00152 is regulated by histone lactylation and promotes cancer cells invasion and migration. Frontiers in Cellular and Infection Microbiology, 12.

[103]. Yang, J., et al. (2022). A positive feedback loop between inactive VHL-triggered histone lactylation and PDGFRβ signaling drives clear cell renal cell carcinoma progression. International Journal of Biological Sciences, 18, 3470–3483.

[104]. Yoon, S., et al. (2021). Circulation, 143, 1912–1925.

[105]. Yu, J., et al. (2021). Histone lactylation drives oncogenesis by facilitating m6A reader protein YTHDF2 expression in ocular melanoma. Genome Biology, 22.

Cite this article

Li,J. (2024). What is the impact of histone modification-based treatment in cardiovascular diseases in modern medicine?. Journal of Food Science, Nutrition and Health,2,40-53.

Data availability

The datasets used and/or analyzed during the current study will be available from the authors upon reasonable request.

Disclaimer/Publisher's Note

The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of EWA Publishing and/or the editor(s). EWA Publishing and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

About volume

Journal:Journal of Food Science, Nutrition and Health

Volume number: Vol.2
ISSN:3029-0821(Print) / 3029-083X(Online)

© 2024 by the author(s). Licensee EWA Publishing, Oxford, UK. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license. Authors who publish this series agree to the following terms:
1. Authors retain copyright and grant the series right of first publication with the work simultaneously licensed under a Creative Commons Attribution License that allows others to share the work with an acknowledgment of the work's authorship and initial publication in this series.
2. Authors are able to enter into separate, additional contractual arrangements for the non-exclusive distribution of the series's published version of the work (e.g., post it to an institutional repository or publish it in a book), with an acknowledgment of its initial publication in this series.
3. Authors are permitted and encouraged to post their work online (e.g., in institutional repositories or on their website) prior to and during the submission process, as it can lead to productive exchanges, as well as earlier and greater citation of published work (See Open access policy for details).