top of page

Changing 2% of Your Genes with Sodium Butyrate: My Second Favorite Salt

To watch the companion video to this blog post, click here.


BACKGROUND ON HISTONE ACETYLATION


Histones are proteins around which long strands of DNA are packaged tightly in a cell nucleus, producing what are called nucleosomes, which aggregate into the chromatin DNA and protein complexes. Histones can be modified at certain positions by methylation, phosphorylation, ubiquitination, SUMOylation, citrullination, and acetylation. Histones 3 and 4 have long tails which are frequently modified, while histones 2A and 2B are often modified at their cores. For example, lysine acetylation of a histone produces more accessible chromatin and is associated with more active transcription.


Histone acetylation is thus part of epigenetic modification. Histone acetyltransferases (HAT) are a family of enzyme that transfer a histone from one molecule (e.g. acetyl coenzyme A) to another (e.g. a lysine residue on the N-terminal tail of a histone). On the other hand, histone deacetylases (HDAC) do the opposite. Generally, acetylation of histones produces a more relaxed chromatin (called a euchromatin) that is associated with greater gene transcription.


There are four classes of HDACs. The first class includes the closely related HDAC1 and HDAC2 and the related HDAC3 and HDAC 8. Class IIA includes HDAC4, HDAC5, HDAC7, and HDAC9. Class IIb includes the similar HDAC6 and HDAC10. Class III’s members are the seven mammalian sirtuins. Class IV includes HDAC11[1].

HDACs are overexpressed in cancerous tumors[2]. Since 2006, the HDAC inhibitor (HDACi) Vorinostat has been FDA approved for the treatment of a kind of lymphoma[3]. HDACis are being researched to attenuate the pathologic epigenetic modifications that accompany cocaine addiction[4], amphetamine addiction[5], and alcohol addiction[6].


MY SUPPLEMENT CHOICE


Designs for Health Tri-Butyrin Supreme ($0.75/300mg softgel). Click here to use my Amazon affiliate link.


MICROBIOME


1. Short-chain fatty acids are a major metabolite of microbial anaerobic fermentation in the human gut. The major metabolites are acetate (C2), propionate (C3), and butyrate (C4), (at a ratio of 60:20:20 in the colon) though there are also branched short-chain fatty acids such as iso-butyrate, valerate, and iso-valerate.

a. Butyrate, whose name is derived from the Greek word for butter, smells like butter and is generated when triglycerides in milk fat experience lipase-catalyzed hydrolysis.

b. Apparently, it responsible for the smell of humans, including their vomit and sweat.

c. It has two isoforms: n-butyric acid and iso-butyric acid, with the former being more plentiful in humans. Both are produced by two different processes in the gut.

d. Under normal conditions, butyrate is only produced from fiber in the gut and circulates the blood in micromolar concentrations. The concentration in blood appears to mostly be low due to metabolism both in the gut and in the liver. Concentration may vary widely across time, with higher peaks.

e. Butyrate is produced by several unrelated species of bacteria, indicating that the process for its metabolism likely developed several times in history.

f. Butyrate content in human feces varies by a factor of 10x.

2. Gut expression of the transporters of butyrate, MCT1 and SMCT1, is governed by NFkB signaling[7]. These transporters are also found in the brain[8].

3. High dosed supplemental sodium butyrate improves gut microbiome structure to the benefit of the host[9].


METABOLISM


1. Butyrate is metabolized quickly, though the prodrug tributyrin delays the metabolism[10][11].

2. Butyrate may increase expression of the structurally similar D-beta-hydroxybutyrate (DHB)[12].


RECEPTOR BINDING


1. Butyrate has been shown to bind to the FFAR2, FFAR3 (both activated by all major SCFAs[13]), HCA2 (also activated by monomethyl fumarate[14] and niacin[15]), and OR51E1 (activated by valeric acids and nonanoic acid[16]) receptors. The OR51E1 receptor may be involved in SCFAs’ influence on the release of serotonin from enterochromaffin cells in the gut.

a. FFAR3[17] and HCA2[18] expression is regulated by DNA methylation. FFAR3 expression is increased in the obese.


HISTONE DEACETYLASE INHIBITION


1. Butyrate inhibits histone deacetylation, which can influence up to 2% of the expression of mammalian genes[19]. It preferentially inhibits classes I and IIa[20][21], and it is the strongest endogenous inhibitor[22].

2. Beta hydroxybutyrate is also an HDACi[23], and it upregulates BDNF due to inhibition of HDAC2 and HDAC3[24].


THE BRAIN


1. An early study found that elevating histone acetylation using butyrate improved both long-term potentiation (LTP) and contextual fear memory in rodents[25].

2. Butyrate has been shown to enhance LTP in several studies[26][27][28][29][30].

3. Butyrate is neuroprotective in models of Huntington’s disease[31][32], ALS[33] (with human trials[34]), Parkinson’s disease[35] (and dopaminergic toxins[36][37]), and vascular dementia[38].

4. Butyrate improves cognitive function in rodent models of Alzheimer’s disease[39] and may reduce beta amyloid plaque levels[40].

5. Butyrate produces neurogenesis in the ischemic rodent brain[41] and is neuroprotective in models of ischemic stroke[42][43].

6. In a rodent model of lipopolysaccharide-induced depression, sodium butyrate reduced activation of the miroglia and improved behavioral symptoms[44].

7. Butyrate increases the neurological effects due to cocaine[45] and amphetamines[46]. It also increases their drug-induced neuroplasticity[47][48][49][50].

8. Butyrate’s plasticity-inducing epigenetic effects on the brain are comparable to that of exercise[51].

9. Sodium butyrate’s effect on neurological disorders appears to be partially mediated by an increased transcription of BDNF due to HDAC inhibition. It appears that HDAC2 and HDAC3 (of Class I) are most relevant to the increased expression of BDNF, whereas HDAC1 is unrelated[52].


BLOOD-BRAIN BARRIER


1. Butyrate insufficiency appears to produce a more permeable blood-brain barrier[53].

2. Butyrate is neuroprotective in rodent models of traumatic brain injury by improving blood-brain barrier quality[54].


DEPRESSION


1. Electroconvulsive shock therapy increases histone H3 acetylation[55]. Deacetylation of hippocampal histones inhibits the antidepressant effect of imipramine in rodents exposed to chronic stress[56]. Sodium butyrate produces an anti-depressant effect on its own and enhances the antidepressant effect of fluoxetine in rodents, likely due to HDAC inhibition and changes in BDNF expression[57]. Similar findings are reported elsewhere[58], though evidence that HDACi is insufficient to produce an antidepressant phenotype has also been produced[59].

2. Butyrate alters epigenetic information on the BDNF gene, causing its overexpression[60].


METABOLIC SYNDROME, LIVER, & T2D


1. Sodium butyrate attenuates hepatic steatosis due to a high-fat diet in rodents[61] and due to a high fat and fructose diet[62].

2. Sodium butyrate improves the inflammatory state in rodent models of T2D[63].

3. Sodium butyrate attenuates diabetic nephropathy by activating Nrf2[64].

4. It improves glycogen metabolism in the livers of diabetic rodents[65].

5. Fortuitously, butyrate protects livers from valproate-induced damage[66].

6. Sodium butyrate improved symptoms of T2D comparably to metformin[67].

7. Sodium butyrate upregulates PPAR gamma in obese rodents[68].


KIDNEYS


1. In a rodent model of kidney disease, butyrate improved renal condition and insulin resistance[69].


CARDIOVASCULAR DISEASE


1. Butyrate reduces oxidative stress at atherosclerotic sites[70].

2. In animal models of high-fat diet-induced atherosclerosis, butyrate reduced plaque development[71].


AUTOIMMUNE DISEASE


1. Sodium butyrate has been shown to improve symptoms of colitis in animals[72], potentially because of improved mucosal synthesis[73] or improvement to gut barrier quality[74].

2. Sodium butyrate may improve symptoms of autoimmune skin disorders[75].

3. In a rodent model of colitis, sodium butyrate improved symptoms by inhibiting NFkB[76].


CANCER


1. Sodium butyrate induces apoptosis in colon[77], rectal[78], and colorectal[79] cancer cell lines. It also does this in breast cancer cells[80]

2. Given sodium butyrate, colorectal cancer cells experience enhanced autophagy due to AMPK (and LKB1) signaling[81].

3. There is a US patent held by Chinese on a method of treating cancer with metformin and sodium butyrate[82].


LONGEVITY


1. Butyrate may increase Foxo3a expression[83].

[1] Witt, O., Deubzer, H. E., Milde, T., & Oehme, I. (2009). HDAC family: What are the cancer relevant targets?. Cancer letters, 277(1), 8-21. [2] Li, Y., & Seto, E. (2016). HDACs and HDAC inhibitors in cancer development and therapy. Cold Spring Harbor perspectives in medicine, 6(10), a026831. [3] Grant, S., Easley, C., & Kirkpatrick, P. (2007). Vorinostat. [4] J Kennedy, P., & Harvey, E. (2015). Histone deacetylases as potential targets for cocaine addiction. CNS & Neurological Disorders-Drug Targets (Formerly Current Drug Targets-CNS & Neurological Disorders), 14(6), 764-772. [5] Arndt, D. L. (2016). Role of HDAC inhibition and environmental condition in altering phases of amphetamine self-administration (Doctoral dissertation, Kansas State University). [6] Bourguet, E., Ozdarska, K., Moroy, G., Jeanblanc, J., & Naassila, M. (2017). Class I HDAC inhibitors: potential new epigenetic therapeutics for alcohol use disorder (AUD). Journal of medicinal chemistry, 61(5), 1745-1766. [7] Borthakur, A., Saksena, S., Gill, R. K., Alrefai, W. A., Ramaswamy, K., & Dudeja, P. K. (2008). Regulation of monocarboxylate transporter 1 (MCT1) promoter by butyrate in human intestinal epithelial cells: Involvement of NF‐κB pathway. Journal of cellular biochemistry, 103(5), 1452-1463. [8] Vijay, N., & Morris, M. E. (2014). Role of monocarboxylate transporters in drug delivery to the brain. Current pharmaceutical design, 20(10), 1487-1498. [9] Wu, W., Xiao, Z., An, W., Dong, Y., & Zhang, B. (2018). Dietary sodium butyrate improves intestinal development and function by modulating the microbial community in broilers. PloS one, 13(5), e0197762. [10] Egorin, M. J., Yuan, Z. M., Sentz, D. L., Plaisance, K., & Eiseman, J. L. (1999). Plasma pharmacokinetics of butyrate after intravenous administration of sodium butyrate or oral administration of tributyrin or sodium butyrate to mice and rats. Cancer chemotherapy and pharmacology, 43(6), 445-453. [11] Miyoshi, M., Sakaki, H., Usami, M., Iizuka, N., Shuno, K., Aoyama, M., & Usami, Y. (2011). Oral administration of tributyrin increases concentration of butyrate in the portal vein and prevents lipopolysaccharide-induced liver injury in rats. Clinical nutrition, 30(2), 252-258. [12] Iriki, T., Tamura, K., Ishii, M., Tanaka, H., Miyamoto, T., & Onda, K. (2009). Concentrations of ketone body and antidiuretic hormone in cerebrospinal fluid in response to the intra‐ruminal administration of butyrate in suckling calves. Animal Science Journal, 80(6), 655-661. [13] Ulven, T. (2012). Short-chain free fatty acid receptors FFA2/GPR43 and FFA3/GPR41 as new potential therapeutic targets. Frontiers in endocrinology, 3, 111. [14] Tang, H., Lu, J. Y. L., Zheng, X., Yang, Y., & Reagan, J. D. (2008). The psoriasis drug monomethylfumarate is a potent nicotinic acid receptor agonist. Biochemical and biophysical research communications, 375(4), 562-565. [15] Singh, N., Gurav, A., Sivaprakasam, S., Brady, E., Padia, R., Shi, H., ... & Lee, J. R. (2014). Activation of Gpr109a, receptor for niacin and the commensal metabolite butyrate, suppresses colonic inflammation and carcinogenesis. Immunity, 40(1), 128-139. [16] Priori, D., Colombo, M., Clavenzani, P., Jansman, A. J., Lallès, J. P., Trevisi, P., & Bosi, P. (2015). The olfactory receptor OR51E1 is present along the gastrointestinal tract of pigs, co-localizes with enteroendocrine cells and is modulated by intestinal microbiota. PLoS One, 10(6), e0129501. [17] Remely, M., Aumueller, E., Merold, C., Dworzak, S., Hippe, B., Zanner, J., ... & Haslberger, A. G. (2014). Effects of short chain fatty acid producing bacteria on epigenetic regulation of FFAR3 in type 2 diabetes and obesity. Gene, 537(1), 85-92. [18] Thangaraju, M., Cresci, G. A., Liu, K., Ananth, S., Gnanaprakasam, J. P., Browning, D. D., ... & Prasad, P. D. (2009). GPR109A is a G-protein–coupled receptor for the bacterial fermentation product butyrate and functions as a tumor suppressor in colon. Cancer research, 69(7), 2826-2832. [19] Davie, J. R. (2003). Inhibition of histone deacetylase activity by butyrate. The Journal of nutrition, 133(7), 2485S-2493S. [20] Davie, J. R. (2003). Inhibition of histone deacetylase activity by butyrate. The Journal of nutrition, 133(7), 2485S-2493S. [21] Cleophas, M. C., Crişan, T. O., Lemmers, H., Toenhake-Dijkstra, H., Fossati, G., Jansen, T. L., ... & Joosten, L. A. (2016). Suppression of monosodium urate crystal-induced cytokine production by butyrate is mediated by the inhibition of class I histone deacetylases. Annals of the rheumatic diseases, 75(3), 593-600. [22] Gilbert, K. M., DeLoose, A., Valentine, J. L., & Fifer, E. K. (2006). Structure–activity relationship between carboxylic acids and T cell cycle blockade. Life sciences, 78(19), 2159-2165. [23] Shimazu, T., Hirschey, M. D., Newman, J., He, W., Shirakawa, K., Le Moan, N., ... & Newgard, C. B. (2013). Suppression of oxidative stress by β-hydroxybutyrate, an endogenous histone deacetylase inhibitor. Science, 339(6116), 211-214. [24] Sleiman, S. F., Henry, J., Al-Haddad, R., El Hayek, L., Abou Haidar, E., Stringer, T., ... & Ninan, I. (2016). Exercise promotes the expression of brain derived neurotrophic factor (BDNF) through the action of the ketone body β-hydroxybutyrate. Elife, 5, e15092. [25] Levenson, J. M., O'Riordan, K. J., Brown, K. D., Trinh, M. A., Molfese, D. L., & Sweatt, J. D. (2004). Regulation of histone acetylation during memory formation in the hippocampus. Journal of Biological Chemistry, 279(39), 40545-40559. [26] Lattal, K. M., Barrett, R. M., & Wood, M. A. (2007). Systemic or intrahippocampal delivery of histone deacetylase inhibitors facilitates fear extinction. Behavioral neuroscience, 121(5), 1125. [27] Vecsey, C. G., Hawk, J. D., Lattal, K. M., Stein, J. M., Fabian, S. A., Attner, M. A., ... & Wood, M. A. (2007). Histone deacetylase inhibitors enhance memory and synaptic plasticity via CREB: CBP-dependent transcriptional activation. Journal of Neuroscience, 27(23), 6128-6140. [28] Haettig, J., Stefanko, D. P., Multani, M. L., Figueroa, D. X., McQuown, S. C., & Wood, M. A. (2011). HDAC inhibition modulates hippocampus-dependent long-term memory for object location in a CBP-dependent manner. Learning & Memory, 18(2), 71-79. [29] Intlekofer, K. A., Berchtold, N. C., Malvaez, M., Carlos, A. J., McQuown, S. C., Cunningham, M. J., ... & Cotman, C. W. (2013). Exercise and sodium butyrate transform a subthreshold learning event into long-term memory via a brain-derived neurotrophic factor-dependent mechanism. Neuropsychopharmacology, 38(10), 2027-2034. [30] Fischer, A., Sananbenesi, F., Wang, X., Dobbin, M., & Tsai, L. H. (2007). Recovery of learning and memory is associated with chromatin remodelling. Nature, 447(7141), 178-182. [31] Gardian, G., Browne, S. E., Choi, D. K., Klivenyi, P., Gregorio, J., Kubilus, J. K., ... & Beal, M. F. (2005). Neuroprotective effects of phenylbutyrate in the N171-82Q transgenic mouse model of Huntington's disease. Journal of Biological Chemistry, 280(1), 556-563. [32] Ferrante, R. J., Kubilus, J. K., Lee, J., Ryu, H., Beesen, A., Zucker, B., ... & Hersch, S. M. (2003). Histone deacetylase inhibition by sodium butyrate chemotherapy ameliorates the neurodegenerative phenotype in Huntington's disease mice. Journal of Neuroscience, 23(28), 9418-9427. [33] Ryu, H., Smith, K., Camelo, S. I., Carreras, I., Lee, J., Iglesias, A. H., ... & Ferrante, R. J. (2005). Sodium phenylbutyrate prolongs survival and regulates expression of anti‐apoptotic genes in transgenic amyotrophic lateral sclerosis mice. Journal of neurochemistry, 93(5), 1087-1098. [34] Cudkowicz, M. E., Andres, P. L., Macdonald, S. A., Bedlack, R. S., Choudry, R., Brown Jr, R. H., ... & Matson, W. R. (2009). Phase 2 study of sodium phenylbutyrate in ALS. Amyotrophic Lateral Sclerosis, 10(2), 99-106. [35] Liu, J., Wang, F., Liu, S., Du, J., Hu, X., Xiong, J., ... & Sun, J. (2017). Sodium butyrate exerts protective effect against Parkinson's disease in mice via stimulation of glucagon like peptide-1. Journal of the neurological sciences, 381, 176-181. [36] Sharma, S., Taliyan, R., & Singh, S. (2015). Beneficial effects of sodium butyrate in 6-OHDA induced neurotoxicity and behavioral abnormalities: Modulation of histone deacetylase activity. Behavioural brain research, 291, 306-314. [37] Gardian, G., Yang, L., Cleren, C., Calingasan, N. Y., Klivenyi, P., & Beal, M. F. (2004). Neuroprotective effects of phenylbutyrate against MPTP neurotoxicity. Neuromolecular medicine, 5(3), 235-241. [38] Liu, J., Sun, J., Wang, F., Yu, X., Ling, Z., Li, H., ... & Yu, J. (2015). Neuroprotective effects of Clostridium butyricum against vascular dementia in mice via metabolic butyrate. BioMed research international, 2015. [39] Govindarajan, N., Agis-Balboa, R. C., Walter, J., Sananbenesi, F., & Fischer, A. (2011). Sodium butyrate improves memory function in an Alzheimer's disease mouse model when administered at an advanced stage of disease progression. Journal of Alzheimer's Disease, 26(1), 187-197. [40] Fernando, W. M. A. D., Martins, I. J., Morici, M., Bharadwaj, P., Rainey-Smith, S. R., Lim, W. L. F., & Martins, R. N. (2020). Sodium butyrate reduces brain amyloid-β levels and improves cognitive memory performance in an Alzheimer’s disease transgenic mouse model at an early disease stage. Journal of Alzheimer's Disease, (Preprint), 1-9. [41] Kim, H. J., Leeds, P., & Chuang, D. M. (2009). The HDAC inhibitor, sodium butyrate, stimulates neurogenesis in the ischemic brain. Journal of neurochemistry, 110(4), 1226-1240. [42] Patnala, R., Arumugam, T. V., Gupta, N., & Dheen, S. T. (2017). HDAC inhibitor sodium butyrate-mediated epigenetic regulation enhances neuroprotective function of microglia during ischemic stroke. Molecular Neurobiology, 54(8), 6391-6411. [43] Park, M. J., & Sohrabji, F. (2016). The histone deacetylase inhibitor, sodium butyrate, exhibits neuroprotective effects for ischemic stroke in middle-aged female rats. Journal of neuroinflammation, 13(1), 1-14. [44] Yamawaki, Y., Yoshioka, N., Nozaki, K., Ito, H., Oda, K., Harada, K., ... & Kanematsu, T. (2018). Sodium butyrate abolishes lipopolysaccharide-induced depression-like behaviors and hippocampal microglial activation in mice. Brain research, 1680, 13-38. [45] Febo, M., Akbarian, S., Schroeder, F. A., & Ferris, C. F. (2009). Cocaine-induced metabolic activation in cortico-limbic circuitry is increased after exposure to the histone deacetylase inhibitor, sodium butyrate. Neuroscience letters, 465(3), 267-271. [46] Kalda, A., Heidmets, L. T., Shen, H. Y., Zharkovsky, A., & Chen, J. F. (2007). Histone deacetylase inhibitors modulates the induction and expression of amphetamine-induced behavioral sensitization partially through an associated learning of the environment in mice. Behavioural brain research, 181(1), 76-84. [47] Kumar, A., Choi, K. H., Renthal, W., Tsankova, N. M., Theobald, D. E., Truong, H. T., ... & Neve, R. L. (2005). Chromatin remodeling is a key mechanism underlying cocaine-induced plasticity in striatum. Neuron, 48(2), 303-314. [48] Sanchis-Segura, C., Lopez-Atalaya, J. P., & Barco, A. (2009). Selective boosting of transcriptional and behavioral responses to drugs of abuse by histone deacetylase inhibition. Neuropsychopharmacology, 34(13), 2642-2654. [49] Schroeder, F. A., Penta, K. L., Matevossian, A., Jones, S. R., Konradi, C., Tapper, A. R., & Akbarian, S. (2008). Drug-induced activation of dopamine D 1 receptor signaling and inhibition of class I/II histone deacetylase induce chromatin remodeling in reward circuitry and modulate cocaine-related behaviors. Neuropsychopharmacology, 33(12), 2981-2992. [50] Shen, H. Y., Kalda, A., Yu, L., Ferrara, J., Zhu, J., & Chen, J. F. (2008). Additive effects of histone deacetylase inhibitors and amphetamine on histone H4 acetylation, cAMP responsive element binding protein phosphorylation and ΔFosB expression in the striatum and locomotor sensitization in mice. Neuroscience, 157(3), 644-655. [51] Intlekofer, K. A., Berchtold, N. C., Malvaez, M., Carlos, A. J., McQuown, S. C., Cunningham, M. J., ... & Cotman, C. W. (2013). Exercise and sodium butyrate transform a subthreshold learning event into long-term memory via a brain-derived neurotrophic factor-dependent mechanism. Neuropsychopharmacology, 38(10), 2027-2034. [52] Sartor, G. C., Malvezzi, A. M., Kumar, A., Andrade, N. S., Wiedner, H. J., Vilca, S. J., ... & Brown, P. T. (2019). Enhancement of BDNF expression and memory by HDAC inhibition requires BET bromodomain reader proteins. Journal of Neuroscience, 39(4), 612-626. [53] Braniste, V., Al-Asmakh, M., Kowal, C., Anuar, F., Abbaspour, A., Tóth, M., ... & Gulyás, B. (2014). The gut microbiota influences blood-brain barrier permeability in mice. Science translational medicine, 6(263), 263ra158-263ra158. [54] Li, H., Sun, J., Wang, F., Ding, G., Chen, W., Fang, R., ... & Liu, J. (2016). Sodium butyrate exerts neuroprotective effects by restoring the blood-brain barrier in traumatic brain injury mice. Brain research, 1642, 70-78. [55] Tsankova, N. M., Kumar, A., & Nestler, E. J. (2004). Histone modifications at gene promoter regions in rat hippocampus after acute and chronic electroconvulsive seizures. Journal of Neuroscience, 24(24), 5603-5610. [56] Tsankova, N. M., Berton, O., Renthal, W., Kumar, A., Neve, R. L., & Nestler, E. J. (2006). Sustained hippocampal chromatin regulation in a mouse model of depression and antidepressant action. Nature neuroscience, 9(4), 519-525. [57] Schroeder, F. A., Lin, C. L., Crusio, W. E., & Akbarian, S. (2007). Antidepressant-like effects of the histone deacetylase inhibitor, sodium butyrate, in the mouse. Biological psychiatry, 62(1), 55-64. [58] Han, A., Sung, Y. B., Chung, S. Y., & Kwon, M. S. (2014). Possible additional antidepressant-like mechanism of sodium butyrate: targeting the hippocampus. Neuropharmacology, 81, 292-302. [59] Yamawaki, Y., Fuchikami, M., Morinobu, S., Segawa, M., Matsumoto, T., & Yamawaki, S. (2012). Antidepressant-like effect of sodium butyrate (HDAC inhibitor) and its molecular mechanism of action in the rat hippocampus. The World Journal of Biological Psychiatry, 13(6), 458-467. [60] Wei, Y. B., Melas, P. A., Wegener, G., Mathé, A. A., & Lavebratt, C. (2015). Antidepressant-like effect of sodium butyrate is associated with an increase in TET1 and in 5-hydroxymethylation levels in the Bdnf gene. International Journal of Neuropsychopharmacology, 18(2). [61] Da Zhou, Q. P., Xin, F. Z., Zhang, R. N., He, C. X., Chen, G. Y., Liu, C., ... & Fan, J. G. (2017). Sodium butyrate attenuates high-fat diet-induced steatohepatitis in mice by improving gut microbiota and gastrointestinal barrier. World Journal of Gastroenterology, 23(1), 60. [62] Baumann, A., Jin, C. J., Brandt, A., Sellmann, C., Nier, A., Burkard, M., ... & Bergheim, I. (2020). Oral supplementation of sodium butyrate attenuates the progression of non-alcoholic steatohepatitis. Nutrients, 12(4), 951. [63] Xu, Y. H., Gao, C. L., Guo, H. L., Zhang, W. Q., Huang, W., Tang, S. S., ... & Zhu, Q. (2018). Sodium butyrate supplementation ameliorates diabetic inflammation in db/db mice. Journal of Endocrinology, 238(3), 231-244. [64] Dong, W., Jia, Y., Liu, X., Zhang, H., Li, T., Huang, W., ... & Wu, H. (2017). Sodium butyrate activates NRF2 to ameliorate diabetic nephropathy possibly via inhibition of HDAC. The Journal of Endocrinology, 232(1), 71. [65] Zhang, W. Q., Zhao, T. T., Gui, D. K., Gao, C. L., Gu, J. L., Gan, W. J., ... & Liu, Z. L. (2019). Sodium butyrate improves liver glycogen metabolism in type 2 diabetes mellitus. Journal of agricultural and food chemistry, 67(27), 7694-7705. [66] Pirozzi, C., Lama, A., Annunziata, C., Cavaliere, G., De Caro, C., Citraro, R., ... & Mollica, M. P. (2020). Butyrate prevents valproate‐induced liver injury: In vitro and in vivo evidence. The FASEB Journal, 34(1), 676-690. [67] Khan, S., & Jena, G. (2016). Sodium butyrate reduces insulin-resistance, fat accumulation and dyslipidemia in type-2 diabetic rat: a comparative study with metformin. Chemico-biological interactions, 254, 124-134. [68] Aguilar, E. C., da Silva, J. F., Navia-Pelaez, J. M., Leonel, A. J., Lopes, L. G., Menezes-Garcia, Z., ... & Alvarez-Leite, J. I. (2018). Sodium butyrate modulates adipocyte expansion, adipogenesis, and insulin receptor signaling by upregulation of PPAR-γ in obese Apo E knockout mice. Nutrition, 47, 75-82. [69] Gonzalez, A., Krieg, R., Massey, H. D., Carl, D., Ghosh, S., Gehr, T. W., & Ghosh, S. S. (2019). Sodium butyrate ameliorates insulin resistance and renal failure in CKD rats by modulating intestinal permeability and mucin expression. Nephrology Dialysis Transplantation, 34(5), 783-794. [70] Aguilar, E. C., dos Santos, L. C., Leonel, A. J., de Oliveira, J. S., Santos, E. A., Navia-Pelaez, J. M., ... & Lemos, V. S. (2016). Oral butyrate reduces oxidative stress in atherosclerotic lesion sites by a mechanism involving NADPH oxidase down-regulation in endothelial cells. The Journal of nutritional biochemistry, 34, 99-105. [71] Du, Y., Li, X., Su, C., Xi, M., Zhang, X., Jiang, Z., ... & Hong, B. (2020). Butyrate protects against high‐fat diet‐induced atherosclerosis via up‐regulating ABCA1 expression in apolipoprotein E‐deficiency mice. British Journal of Pharmacology, 177(8), 1754-1772. [72] Vieira, E. L., Leonel, A. J., Sad, A. P., Beltrão, N. R., Costa, T. F., Ferreira, T. M., ... & Alvarez-Leite, J. I. (2012). Oral administration of sodium butyrate attenuates inflammation and mucosal lesion in experimental acute ulcerative colitis. The Journal of nutritional biochemistry, 23(5), 430-436. [73] Finnie, I. A., Dwarakanath, A. D., Taylor, B. A., & Rhodes, J. M. (1995). Colonic mucin synthesis is increased by sodium butyrate. Gut, 36(1), 93-99. [74] Chen, G., Ran, X., Li, B., Li, Y., He, D., Huang, B., ... & Wang, W. (2018). Sodium butyrate inhibits inflammation and maintains epithelium barrier integrity in a TNBS-induced inflammatory bowel disease mice model. EBioMedicine, 30, 317-325. [75] Schwarz, A., Bruhs, A., & Schwarz, T. (2017). The short-chain fatty acid sodium butyrate functions as a regulator of the skin immune system. Journal of Investigative Dermatology, 137(4), 855-864. [76] Lee, C., Kim, B. G., Kim, J. H., Chun, J., Im, J. P., & Kim, J. S. (2017). Sodium butyrate inhibits the NF-kappa B signaling pathway and histone deacetylation, and attenuates experimental colitis in an IL-10 independent manner. International immunopharmacology, 51, 47-56. [77] Hague, A., Manning, A. M., Hanlon, K. A., Hart, D., Paraskeva, C., & Huschtscha, L. I. (1993). Sodium butyrate induces apoptosis in human colonic tumour cell lines in a p53‐independent pathway: implications for the possible role of dietary fibre in the prevention of large‐bowel cancer. International journal of cancer, 55(3), 498-505. [78] Verma, S. P., Agarwal, A., & Das, P. (2018). Sodium butyrate induces cell death by autophagy and reactivates a tumor suppressor gene DIRAS1 in renal cell carcinoma cell line UOK146. In Vitro Cellular & Developmental Biology-Animal, 54(4), 295-303. [79] Wang, W., Fang, D., Zhang, H., Xue, J., Wangchuk, D., Du, J., & Jiang, L. (2020). Sodium Butyrate Selectively Kills Cancer Cells and Inhibits Migration in Colorectal Cancer by Targeting Thioredoxin-1. OncoTargets and Therapy, 13, 4691-4704. [80] Salimi, V., Shahsavari, Z., Safizadeh, B., Hosseini, A., Khademian, N., & Tavakoli-Yaraki, M. (2017). Sodium butyrate promotes apoptosis in breast cancer cells through reactive oxygen species (ROS) formation and mitochondrial impairment. Lipids in health and disease, 16(1), 1-11. [81] Luo, S., Li, Z., Mao, L., Chen, S., & Sun, S. (2019). Sodium butyrate induces autophagy in colorectal cancer cells through LKB1/AMPK signaling. Journal of physiology and biochemistry, 75(1), 53-63. [82] Kwan-Hwa, C. H. I., Wang, Y. S., & Chang, C. C. (2018). U.S. Patent No. 9,913,816. Washington, DC: U.S. Patent and Trademark Office. [83] Honma, K., Oshima, K., Takami, S., & Goda, T. (2020). Regulation of hepatic genes related to lipid metabolism and antioxidant enzymes by sodium butyrate supplementation. Metabolism Open, 7, 100043.

bottom of page