Skip to main content
Top
Published in: Breast Cancer 4/2020

01-07-2020 | Breast Cancer | Original Article

Comparative effect of sodium butyrate and sodium propionate on proliferation, cell cycle and apoptosis in human breast cancer cells MCF-7

Authors: Josiane Semaan, Sandy El-Hakim, José-Noel Ibrahim, Rémi Safi, Arpiné Ardzivian Elnar, Charbel El Boustany

Published in: Breast Cancer | Issue 4/2020

Login to get access

Abstract

Introduction

Short-chain fatty acids (SCFAs) are ubiquitous lipids produced as a result of bacterial fermentation of dietary fiber. While their role in colorectal cancer is well known, the effect of SCFAs in breast cancer is poorly defined.

Objective

To understand the various effects of SCFAs on breast carcinogenesis, we investigated the effect of sodium butyrate (NaB) and sodium propionate (NaP) in MCF-7 cell line.

Materials and methods

Cells were incubated with different concentrations of NaB or NaP for 24, 48, 72 or 96 h. Cell proliferation was assayed using MTT kit. Cell cycle was performed using propidium iodide staining then analyzed with a flow cytometer. Apoptosis was assessed by Hoechst technique and cell-cycle sub-G1 phase.

Results

NaB and NaP inhibited MCF-7 cell proliferation in a dose-dependent manner with respective IC50 of 1.26 mM and 4.5 mM, thus indicating that NaB is more potent than NaP. Low and medium levels of both SCFAs induced morphology changes which are characteristic of a differentiated phenotype. Flow cytometry analysis revealed a blockage in G1 growth phase. Interestingly, removing NaB or NaP from culture media after few days of treatment showed a reversible effect on cell morphology and proliferation where cells reentered the cycle after 24 h of drug wash-out. Finally, treatment with medium levels of these molecules induced low MCF-7 apoptosis, while higher doses led to massive apoptosis.

Conclusion

Our results show that SCFAs may be considered as an interesting inhibitor for breast cancer progression.
Appendix
Available only for authorised users
Literature
1.
go back to reference Sealy L, Chalkley R. The effect of sodium butyrate on histone modification. Cell. 1978;14:115–21.CrossRef Sealy L, Chalkley R. The effect of sodium butyrate on histone modification. Cell. 1978;14:115–21.CrossRef
2.
go back to reference Davie JR. Inhibition of histone deacetylase activity by butyrate. J Nutr. 2485S;133:2485S–S24932493.CrossRef Davie JR. Inhibition of histone deacetylase activity by butyrate. J Nutr. 2485S;133:2485S–S24932493.CrossRef
3.
go back to reference Wang Y, Hu PC, Ma YB, Fan R, Gao FF, Zhang JW, et al. Sodium butyrate-induced apoptosis and ultrastructural changes in MCF-7 breast cancer cells. Ultrastruct Pathol. 2016;40:200–4.CrossRef Wang Y, Hu PC, Ma YB, Fan R, Gao FF, Zhang JW, et al. Sodium butyrate-induced apoptosis and ultrastructural changes in MCF-7 breast cancer cells. Ultrastruct Pathol. 2016;40:200–4.CrossRef
4.
go back to reference Pant K, Yadav AK, Gupta P, Islam R, Saraya A, Venugopal SK. Butyrate induces ROS-mediated apoptosis by modulating miR-22/SIRT-1 pathway in hepatic cancer cells. Redox Biol. 2017;12:340–9.CrossRef Pant K, Yadav AK, Gupta P, Islam R, Saraya A, Venugopal SK. Butyrate induces ROS-mediated apoptosis by modulating miR-22/SIRT-1 pathway in hepatic cancer cells. Redox Biol. 2017;12:340–9.CrossRef
5.
go back to reference Mahadevan V. HDAC inhibitors show differential epigenetic regulation and cell survival strategies on p53 mutant colon cancer cells. J Biomol Struct Dyn. 2018;36:938–55.CrossRef Mahadevan V. HDAC inhibitors show differential epigenetic regulation and cell survival strategies on p53 mutant colon cancer cells. J Biomol Struct Dyn. 2018;36:938–55.CrossRef
6.
go back to reference Han R, Sun Q, Wu J, Zheng P, Zhao G. Sodium butyrate upregulates miR-203 expression to exert anti-proliferation effect on colorectal cancer cells. Cell Physiol Biochem. 2016;39:1919–29.CrossRef Han R, Sun Q, Wu J, Zheng P, Zhao G. Sodium butyrate upregulates miR-203 expression to exert anti-proliferation effect on colorectal cancer cells. Cell Physiol Biochem. 2016;39:1919–29.CrossRef
7.
go back to reference Ma J, Zhang Y, Wang J, Zhao T, Ji P, Song J, et al. Proliferative effects of gamma-amino butyric acid on oral squamous cell carcinoma cells are associated with mitogen-activated protein kinase signaling pathways. Int J Mol Med. 2016;38:305–11.CrossRef Ma J, Zhang Y, Wang J, Zhao T, Ji P, Song J, et al. Proliferative effects of gamma-amino butyric acid on oral squamous cell carcinoma cells are associated with mitogen-activated protein kinase signaling pathways. Int J Mol Med. 2016;38:305–11.CrossRef
8.
go back to reference Khan MA, Ahmad R, Srivastava AN. Effect of methyl butyrate aroma on the survival and viability of human breast cancer cells in vitro. J Egypt Natl Canc Inst. 2016;28:81–8.CrossRef Khan MA, Ahmad R, Srivastava AN. Effect of methyl butyrate aroma on the survival and viability of human breast cancer cells in vitro. J Egypt Natl Canc Inst. 2016;28:81–8.CrossRef
9.
go back to reference Damaskos C, Garmpis N, Valsami S, Kontos M, Spartalis E, Kalampokas T, et al. Histone deacetylase inhibitors: an attractive therapeutic strategy against breast cancer. Anticancer Res. 2017;37:35–46.CrossRef Damaskos C, Garmpis N, Valsami S, Kontos M, Spartalis E, Kalampokas T, et al. Histone deacetylase inhibitors: an attractive therapeutic strategy against breast cancer. Anticancer Res. 2017;37:35–46.CrossRef
10.
go back to reference Huang W, Ren C, Huang G, Liu J, Liu W, Wang L, et al. Inhibition of store-operated Ca(2+) entry counteracts the apoptosis of nasopharyngeal carcinoma cells induced by sodium butyrate. Oncol Lett. 2017;13:921–9.CrossRef Huang W, Ren C, Huang G, Liu J, Liu W, Wang L, et al. Inhibition of store-operated Ca(2+) entry counteracts the apoptosis of nasopharyngeal carcinoma cells induced by sodium butyrate. Oncol Lett. 2017;13:921–9.CrossRef
11.
go back to reference Abe M, Kufe DW. Effect of sodium butyrate on human breast carcinoma (MCF-7) cellular proliferation, morphology, and CEA production. Breast Cancer Res Treat. 1984;4:269–74.CrossRef Abe M, Kufe DW. Effect of sodium butyrate on human breast carcinoma (MCF-7) cellular proliferation, morphology, and CEA production. Breast Cancer Res Treat. 1984;4:269–74.CrossRef
12.
go back to reference Kim J, Park H, Im JY, Choi WS, Kim HS. Sodium butyrate regulates androgen receptor expression and cell cycle arrest in human prostate cancer cells. Anticancer Res. 2007;27:3285–92.PubMed Kim J, Park H, Im JY, Choi WS, Kim HS. Sodium butyrate regulates androgen receptor expression and cell cycle arrest in human prostate cancer cells. Anticancer Res. 2007;27:3285–92.PubMed
13.
go back to reference Siavoshian S, Blottiere HM, Cherbut C, Galmiche JP. Butyrate stimulates cyclin D and p21 and inhibits cyclin-dependent kinase 2 expression in HT-29 colonic epithelial cells. Biochem Biophys Res Commun. 1997;232:169–72.CrossRef Siavoshian S, Blottiere HM, Cherbut C, Galmiche JP. Butyrate stimulates cyclin D and p21 and inhibits cyclin-dependent kinase 2 expression in HT-29 colonic epithelial cells. Biochem Biophys Res Commun. 1997;232:169–72.CrossRef
14.
go back to reference Glass KA, McDonnell LM, Rassel RC, Zierke KL. Controlling Listeria monocytogenes on sliced ham and turkey products using benzoate, propionate, and sorbate. J Food Prot. 2007;70:2306–12.CrossRef Glass KA, McDonnell LM, Rassel RC, Zierke KL. Controlling Listeria monocytogenes on sliced ham and turkey products using benzoate, propionate, and sorbate. J Food Prot. 2007;70:2306–12.CrossRef
15.
go back to reference Yoon SK, Ahn Y-H. Application of sodium propionate to the suspension culture of Chinese hamster ovary cells for enhanced production of follicle-stimulating hormone. Biotechnol Bioprocess Eng. 2007;12:497.CrossRef Yoon SK, Ahn Y-H. Application of sodium propionate to the suspension culture of Chinese hamster ovary cells for enhanced production of follicle-stimulating hormone. Biotechnol Bioprocess Eng. 2007;12:497.CrossRef
16.
go back to reference El Boustany C, Bidaux G, Enfissi A, Delcourt P, Prevarskaya N, Capiod T. Capacitative calcium entry and transient receptor potential canonical 6 expression control human hepatoma cell proliferation. Hepatology. 2008;47:2068–77.CrossRef El Boustany C, Bidaux G, Enfissi A, Delcourt P, Prevarskaya N, Capiod T. Capacitative calcium entry and transient receptor potential canonical 6 expression control human hepatoma cell proliferation. Hepatology. 2008;47:2068–77.CrossRef
17.
go back to reference El Boustany C, Katsogiannou M, Delcourt P, Dewailly E, Prevarskaya N, Borowiec AS, et al. Differential roles of STIM1, STIM2 and Orai1 in the control of cell proliferation and SOCE amplitude in HEK293 cells. Cell Calcium. 2009;47:350–9.CrossRef El Boustany C, Katsogiannou M, Delcourt P, Dewailly E, Prevarskaya N, Borowiec AS, et al. Differential roles of STIM1, STIM2 and Orai1 in the control of cell proliferation and SOCE amplitude in HEK293 cells. Cell Calcium. 2009;47:350–9.CrossRef
18.
go back to reference Katsogiannou M, El Boustany C, Gackiere F, Delcourt P, Athias A, Mariot P, et al. Caveolae contribute to the apoptosis resistance induced by the alpha(1A)-adrenoceptor in androgen-independent prostate cancer cells. PLoS ONE. 2009;4:e7068.CrossRef Katsogiannou M, El Boustany C, Gackiere F, Delcourt P, Athias A, Mariot P, et al. Caveolae contribute to the apoptosis resistance induced by the alpha(1A)-adrenoceptor in androgen-independent prostate cancer cells. PLoS ONE. 2009;4:e7068.CrossRef
19.
go back to reference Al-Dhaheri M, Wu J, Skliris GP, Li J, Higashimato K, Wang Y, et al. CARM1 is an important determinant of ERalpha-dependent breast cancer cell differentiation and proliferation in breast cancer cells. Cancer Res. 2011;71:2118–28.CrossRef Al-Dhaheri M, Wu J, Skliris GP, Li J, Higashimato K, Wang Y, et al. CARM1 is an important determinant of ERalpha-dependent breast cancer cell differentiation and proliferation in breast cancer cells. Cancer Res. 2011;71:2118–28.CrossRef
20.
go back to reference Zhang Z, Lei A, Xu L, Chen L, Chen Y, Zhang X, et al. Similarity in gene-regulatory networks suggests that cancer cells share characteristics of embryonic neural cells. J Biol Chem. 2017;292:12842–59.CrossRef Zhang Z, Lei A, Xu L, Chen L, Chen Y, Zhang X, et al. Similarity in gene-regulatory networks suggests that cancer cells share characteristics of embryonic neural cells. J Biol Chem. 2017;292:12842–59.CrossRef
21.
go back to reference Cummings BS, Schnellmann RG. Measurement of cell death in mammalian cells. Curr Protoc Pharmacol. 2004;12:12–8. Cummings BS, Schnellmann RG. Measurement of cell death in mammalian cells. Curr Protoc Pharmacol. 2004;12:12–8.
22.
go back to reference McMichael AJ, Potter JD. Dietary influences upon colon carcinogenesis. Princess Takamatsu Symp. 1985;16:275–90.PubMed McMichael AJ, Potter JD. Dietary influences upon colon carcinogenesis. Princess Takamatsu Symp. 1985;16:275–90.PubMed
23.
go back to reference Jacobs LR. Relationship between dietary fiber and cancer: metabolic, physiologic, and cellular mechanisms. Proc Soc Exp Biol Med. 1986;183:299–310.CrossRef Jacobs LR. Relationship between dietary fiber and cancer: metabolic, physiologic, and cellular mechanisms. Proc Soc Exp Biol Med. 1986;183:299–310.CrossRef
24.
go back to reference Jacobs LR. Effect of dietary fiber on colonic cell proliferation and its relationship to colon carcinogenesis. Prev Med. 1987;16:566–71.CrossRef Jacobs LR. Effect of dietary fiber on colonic cell proliferation and its relationship to colon carcinogenesis. Prev Med. 1987;16:566–71.CrossRef
25.
go back to reference Jacobs R. Role of dietary factors in cell replication and colon cancer. Am J Clin Nutr. 1988;48:775–9.CrossRef Jacobs R. Role of dietary factors in cell replication and colon cancer. Am J Clin Nutr. 1988;48:775–9.CrossRef
26.
go back to reference Wu X, Wu Y, He L, Wu L, Wang X, Liu Z. Effects of the intestinal microbial metabolite butyrate on the development of colorectal cancer. J Cancer. 2018;9:2510–7.CrossRef Wu X, Wu Y, He L, Wu L, Wang X, Liu Z. Effects of the intestinal microbial metabolite butyrate on the development of colorectal cancer. J Cancer. 2018;9:2510–7.CrossRef
27.
go back to reference Wang G, Yu Y, Wang YZ, Wang JJ, Guan R, Sun Y, et al. Role of SCFAs in gut microbiome and glycolysis for colorectal cancer therapy. J Cell Physiol. 2019;234:17023–49.CrossRef Wang G, Yu Y, Wang YZ, Wang JJ, Guan R, Sun Y, et al. Role of SCFAs in gut microbiome and glycolysis for colorectal cancer therapy. J Cell Physiol. 2019;234:17023–49.CrossRef
28.
go back to reference Hinnebusch BF, Meng S, Wu JT, Archer SY, Hodin RA. The effects of short-chain fatty acids on human colon cancer cell phenotype are associated with histone hyperacetylation. J Nutr. 2002;132:1012–7.CrossRef Hinnebusch BF, Meng S, Wu JT, Archer SY, Hodin RA. The effects of short-chain fatty acids on human colon cancer cell phenotype are associated with histone hyperacetylation. J Nutr. 2002;132:1012–7.CrossRef
29.
go back to reference West AC, Johnstone RW. New and emerging HDAC inhibitors for cancer treatment. J Clin Investig. 2014;124:30–9.CrossRef West AC, Johnstone RW. New and emerging HDAC inhibitors for cancer treatment. J Clin Investig. 2014;124:30–9.CrossRef
30.
go back to reference Goey AK, Sissung TM, Peer CJ, Figg WD. Pharmacogenomics and histone deacetylase inhibitors. Pharmacogenomics. 2016;17:1807–15.CrossRef Goey AK, Sissung TM, Peer CJ, Figg WD. Pharmacogenomics and histone deacetylase inhibitors. Pharmacogenomics. 2016;17:1807–15.CrossRef
31.
go back to reference Eckschlager T, Plch J, Stiborova M, Hrabeta J. Histone deacetylase inhibitors as anticancer drugs. Int J Mol Sci. 2017;18:1414.CrossRef Eckschlager T, Plch J, Stiborova M, Hrabeta J. Histone deacetylase inhibitors as anticancer drugs. Int J Mol Sci. 2017;18:1414.CrossRef
32.
go back to reference Li Y, Peng L, Seto E. Histone deacetylase 10 regulates the cell cycle G2/M phase transition via a novel let-7-HMGA2-cyclin A2 pathway. Mol Cell Biol. 2015;35:3547–65.CrossRef Li Y, Peng L, Seto E. Histone deacetylase 10 regulates the cell cycle G2/M phase transition via a novel let-7-HMGA2-cyclin A2 pathway. Mol Cell Biol. 2015;35:3547–65.CrossRef
33.
go back to reference Jang YG, Hwang KA, Choi KC. Rosmarinic acid, a component of rosemary tea, induced the cell cycle arrest and apoptosis through modulation of HDAC2 expression in prostate cancer cell lines. Nutrients. 2018;10:1784.CrossRef Jang YG, Hwang KA, Choi KC. Rosmarinic acid, a component of rosemary tea, induced the cell cycle arrest and apoptosis through modulation of HDAC2 expression in prostate cancer cell lines. Nutrients. 2018;10:1784.CrossRef
34.
go back to reference Pattayil L, Balakrishnan-Saraswathi HT. In vitro evaluation of apoptotic induction of butyric acid derivatives in colorectal carcinoma cells. Anticancer Res. 2019;39:3795–801.CrossRef Pattayil L, Balakrishnan-Saraswathi HT. In vitro evaluation of apoptotic induction of butyric acid derivatives in colorectal carcinoma cells. Anticancer Res. 2019;39:3795–801.CrossRef
35.
go back to reference Cao M, Zhang Z, Han S, Lu X. Butyrate inhibits the proliferation and induces the apoptosis of colorectal cancer HCT116 cells via the deactivation of mTOR/S6K1 signaling mediated partly by SIRT1 downregulation. Mol Med Rep. 2019;19:3941–7.PubMed Cao M, Zhang Z, Han S, Lu X. Butyrate inhibits the proliferation and induces the apoptosis of colorectal cancer HCT116 cells via the deactivation of mTOR/S6K1 signaling mediated partly by SIRT1 downregulation. Mol Med Rep. 2019;19:3941–7.PubMed
36.
go back to reference Jia X, Zheng Y, Guo Y, Chen K. Sodium butyrate and panobinostat induce apoptosis of chronic myeloid leukemia cells via multiple pathways. Mol Genet Genomic Med. 2019;7:e613.CrossRef Jia X, Zheng Y, Guo Y, Chen K. Sodium butyrate and panobinostat induce apoptosis of chronic myeloid leukemia cells via multiple pathways. Mol Genet Genomic Med. 2019;7:e613.CrossRef
37.
go back to reference Taylor MA, Khathayer F, Ray SK. Quercetin and sodium butyrate synergistically increase apoptosis in rat C6 and human T98G glioblastoma cells through inhibition of autophagy. Neurochem Res. 2019;44:1715–25.CrossRef Taylor MA, Khathayer F, Ray SK. Quercetin and sodium butyrate synergistically increase apoptosis in rat C6 and human T98G glioblastoma cells through inhibition of autophagy. Neurochem Res. 2019;44:1715–25.CrossRef
38.
go back to reference Salimi V, Shahsavari Z, Safizadeh B, Hosseini A, Khademian N, Tavakoli-Yaraki M. Sodium butyrate promotes apoptosis in breast cancer cells through reactive oxygen species (ROS) formation and mitochondrial impairment. Lipids Health Dis. 2017;16:208.CrossRef Salimi V, Shahsavari Z, Safizadeh B, Hosseini A, Khademian N, Tavakoli-Yaraki M. Sodium butyrate promotes apoptosis in breast cancer cells through reactive oxygen species (ROS) formation and mitochondrial impairment. Lipids Health Dis. 2017;16:208.CrossRef
39.
go back to reference Kim K, Kwon O, Ryu TY, Jung CR, Kim J, Min JK, et al. Propionate of a microbiota metabolite induces cell apoptosis and cell cycle arrest in lung cancer. Mol Med Rep. 2019;20:1569–74.PubMedPubMedCentral Kim K, Kwon O, Ryu TY, Jung CR, Kim J, Min JK, et al. Propionate of a microbiota metabolite induces cell apoptosis and cell cycle arrest in lung cancer. Mol Med Rep. 2019;20:1569–74.PubMedPubMedCentral
40.
go back to reference Ryu TY, Kim K, Son MY, Min JK, Kim J, Han TS, et al. Downregulation of PRMT1, a histone arginine methyltransferase, by sodium propionate induces cell apoptosis in colon cancer. Oncol Rep. 2019;41:1691–9.PubMed Ryu TY, Kim K, Son MY, Min JK, Kim J, Han TS, et al. Downregulation of PRMT1, a histone arginine methyltransferase, by sodium propionate induces cell apoptosis in colon cancer. Oncol Rep. 2019;41:1691–9.PubMed
Metadata
Title
Comparative effect of sodium butyrate and sodium propionate on proliferation, cell cycle and apoptosis in human breast cancer cells MCF-7
Authors
Josiane Semaan
Sandy El-Hakim
José-Noel Ibrahim
Rémi Safi
Arpiné Ardzivian Elnar
Charbel El Boustany
Publication date
01-07-2020
Publisher
Springer Japan
Published in
Breast Cancer / Issue 4/2020
Print ISSN: 1340-6868
Electronic ISSN: 1880-4233
DOI
https://doi.org/10.1007/s12282-020-01063-6

Other articles of this Issue 4/2020

Breast Cancer 4/2020 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine