Recently I came upon the epidrug concept, which really grabbed my attention and urged me to learn about this area. As it turns out this may be actually the root of my POIS. This is nothing new of course as others on the forum have already proposed this many years ago and methylation issues were extensively discussed as well. Still I believe this issue was not adequately addressed and mostly undermethylation was considered as a possible source. However studying this problem with more scrutiny I have come to the conclusion that in my case hypermethylation must be the underlying cause. This also makes it clear why I had the most success with anti-cancer supplements and at least partly resolves the similarities with prostate or breast cancer, multiple sclerosis, Alzheimer, COVID-19 and possibly a number of other diseases as well.
The simple notion that POIS may be acquired secondarily should imply of its epigenetic origin at least in a number of cases. I haven't read everything about methylation on the site, but I simply can't understand why the two key factors in this regard were not even mentioned before. These are
DNA methyl transferases (DNMTs) and histone deacetylases (HDACs). DNMTs are classified as maintenance (Dnmt1) and de novo methyltransferases (Dnmt3a, Dnmt3b) and for the time being DNMT1 was the focus of my research. Insufficient methylation can cause problems of its own and could explain why many POISers had success with methylation support. However hypermethylation was mostly the focus of cancer research so far, even though researchers are beginning to realize that it may contribute to the development of many other diseases as well.
Hypermethylation may occur due to prolonged inflammation that upregulates DNMT1 and leads to gene promoter (CpG islands) methylation, which causes specific genes to become inactive or silent. Many of these genes code cancer suppressive functions, so their loss could easily lead to development of malignancy. It is true that I don't have cancer, though I may be prone to it. Nevertheless other genes could be also silenced this way, which could lead to metabolic dysfunction. Most importantly this hypermethylation (gene silencing) could occur tissue or organ specifically which could explain different types of cancer or why our disease is different from CFS/FM or asthma even though particular treatments could apparently have the same benefit. I can't deny the possibility that some POIS start in the brain as some believe, however I am practically convinced that for me it is the genital region and the prostate being the most likely culprit. Hypermethylation may silence both ERB and SRD5A2. As DHT is a precursor of 3B-Adiol this could lead to
both receptor and ligand insufficiency. POIS was also associated with a possible loss of 5a-reductase function which may be caused by hypermethyaltion of prostatic SRD5A2.
Regarding my own case the most enlightening revelation came from a study which concludes that
inhibition of DNMT1 and HDAC lead to the restoration of estrogen receptor beta function and expression. What is even better the combination of such inhibitors have an even greater effect and this was actually proven on prostate cancer cells as an addition. I think it is needless to say how this translates to my own particular case given my success with ERB agonists. Gene silencing may also impair DNA repair and hormonal response and the involvement of 3beta-Adiol is even implied in this study. ERB itself upregulates quinone reductase and glutathione-S-reductase thereby confers anti-oxidative support. It is also proposed that ERB may antagonize the proliferative and pro-oxidant actions of both ERa and AR. This could be interesting if we consider that ERa antagonists (e.g. artemisinin) or AR antagonists (e.g. silodosin) also proved useful for some POISer, though I also had success with testosterone boosters.
Histone acetylation could be also targeted as it can activate transcription.
The re-expression of ERB probably involves the upregulation of E-cadherin expression.http://irep.ntu.ac.uk/id/eprint/22538/1/192797_1177%20Mcardle%20PostPrint.pdfResveratrol can inhibit both DNMT1 and HDAC1 which protects against breast cancer. In this respect synergistic effects were seen with melatonin, tea polyphenols (EGCG), quercetin and gamma-tocotrienol.
https://pdfs.nutramedix.ec/Reveratrol%20-%20Cancer%20(preclinical).pdfThrough SIRT-1 activation resveratrol may decrease viral replication and hyper-inflammation in viral infections.
It is proposed that antiviral drugs could be combined with epidrugs (DNMTi and HDACi) combating viral infections like SARS-CoV. For example, various antiviral drugs such as remdesivir, ribavirin, favipiravir, and galidesivir could be combined with DNMT inhibitors such as decitabine, azacitidine or HDAC inhibitors, including vorinostat, belinostat, panobinostat, TSA.
In silico study on the combination of vitamin C, curcumin and glycyrrhizic acid (VCG Plus) revealed strong immunoprotection against SARS-CoV-2 infection by inhibiting the inflammatory response and cytokine storm.https://www.sciencedirect.com/science/article/pii/S0753332221008787SIRT3, a class III HDAC, restricted HBV cccDNA transcription. RNA viruses can also recruit host DNA methyltransferases (DNMTs) to methylate and decrease gene expression of specific genes including those for shaping innate and adaptive immune responses. Inhibition of DNMTs will reactivate the immunity-related gene function responsible for combating viral infection.
The most important compound of Nigella Sativa is Thymoquinone (TQ) which is known for epigenetic action. Thymoquinone can suppress UHRF1 and, thus, might be able to repair epigenetic aberrations in cancer cells through a DNA demethylating process, most likely through the downregulation of DNMT 1.https://www.ffhdj.com/index.php/ffhd/article/view/707/1259 Age-related macular degeneration (AMD) patients exhibited up-regulation of DNMT1 and DNMT3B expression compared to healthy peers.Sirtuin1 (SIRT1) is a NAD+ dependent HDAC, which makes the action of resveratrol perplexing in view of other research. DNMT1 and LINE-1 are positively correlated. LINE-1 methylation was also higher in Alzheimer’s disease (AD) and other similarities were also found with AMD. Resveratrol is also recommended for the treatment for both AMD and AD in different studies.
https://www.mdpi.com/2077-0383/8/2/159/htmIt has to be noted that in human retinal pigment epithelial cells resveratrol had an opposite effect as it increased DNMT1. This contradiction may be due to different tissues examined or a general balancing effect of resveratrol in this regard. Nevertheless resveratrol proved the best treatment for my photophobia so this has to be mentioned. Bacterial LPS may also reduce DNMT1 and LINE-1 in this setting, although as shown later inflammatory processes usually enhance DNMT1.
https://www.mdpi.com/1422-0067/19/7/2118/htmDeletion of DNMT3b gene in a colorectal cancer cell line led to less than 3% loss of global DNA methylation, however concomitant distruption of DNMT1 and DNMT3b reduced genomic DNA methylation by greater than 95%.
https://www.nature.com/articles/416552aS-adenosyl-L-methionine (SAMe) serves as an effective methyl donor while S-adenosyl-L-homocysteine (SAH) is a potent modulator of DNMTs and HNMTs activity. Dietary folic acid (200 mg/day) and vitamin B1 (0.8–1 mg/day) are essential for conversion of homocysteine (hyc) to methionine. Low levels of folic acid and high levels of serum hyc have been related to hypermethylation in patients.Histone tails acetylation mediated by histone acetyltransferases (HATs) leads to euchromatin, whereas histone deacetylation by deacetylases (HDACs) leads to heterochromatin with reduced DNA accessibility. This makes it likely that increasing euchromatin compared to heterochromatin is beneficial for me.
Alpha-ketoglutarate (aKG), succinate and fumarate are products of
acetyl coenzyme A (acetyl-CoA) and a low aKG hampers demethylation. Acetyl-CoA is known to activate HATs.
Barley beta-glucan and beta-hydroxybutyrate inhibit HDAC1 and increases histone acetylation that induces antioxidant genes.
Polyphenols are potent inhibitors of DNMTs and regulate genes encoding important enzymes including sirtuin-type deacetylases and transcription factors in in vitro and in vivo models.
Dietary compounds may regulate HATs (i.e. curcumin, catechin, genistein, diallyl disulfide) and HDACs (i.e. b-glucan, sulforaphane, curcumin, butyrate, resveratrol, quercetin) activity.NAD+ regulates Class III HDACs, also termed sirtuins (Sirts). The sirtuins that rely on NAD+ activity are generally cardioprotective in both animal models and humans. Nicotinamide riboside, a NAD+ precursor abundant in cow milk, stimulates Sirt1 preventing cardiac injury in septic mice and arterial stiffness in older humans.Resveratrol and PUFAs differentially up- and down-regulate miRNAs. Beta-glucan affects gut microbiota composition as it increases
propionate (HDACs inhibitor) producing Bacteriodes while reducing Firmicutes.
The coffee polyphenols (caffeic and chlorogenic acids) are potent inhibitors of DNMTs in humans.Figure 1. Epigenetic_mechanisms
Figure 2. FeedingTheEpigenome
http://statics.drvoice.cn/uploadfile/2019/0217/20190217055754211.pdfSAM is possibly detrimental for me, while SAH could be beneficial as a DNMT inhibitor.
In this mouse model, folate supplementation led to a reduction in tumor numbers if administered prior to tumor development; in contrast, folate deficiency conferred protection after tumor formation. Folate deficiency resulted in a significant decrease in SAM levels in almost all groups. In the group in which the decreased SAM was not statistically significant, a significant increase in SAH was observed. The observed alterations in SAM or SAH are characteristic of folate deficiency and indicate the effectiveness of the folate-deficient diet. It is postulated that lower levels of SAM can result in DNA hypomethylation.https://academic.oup.com/carcin/article/24/1/39/2608348?login=trueCould this be the reason for my problems with zinc?
Sarcosine is an epigenetic modifier that can induce DNA hypermethylation in combination with SAMe excess. Sarcosine did not increase spermidine and spermine in this study, although elsewhere it is indicated that they are positively correlated. Sarcosine is also upregulated in some breast tumours. High intracellular zinc levels could be responsible for sarcosine-induced effects as DNA methylation is depressed in zinc deficiency. Zinc deficiency does not impair the synthesis of SAMe and SAH, but slowed SAMe activity.
https://febs.onlinelibrary.wiley.com/doi/full/10.1002/1878-0261.12439EGCG inhibits DNMT, but increases SAM, which may explain my negative experiences with it. In a different study creatine supplementation also increased SAM, which may associate SAM to my lymphatic issues.
SAM serves as the endogenous co-substrate and binds to the SET domain of EZH2. Although EZH2-mediated methylation also contributes as a potential independent mechanism for epigenetic gene silencing, it also cooperates with other enzymes that participate in epigenetic gene silencing. A physical and functional relationship between EZH2 and DNMTs has been recently documented.
A dose-dependent inhibition of DNMT activity was observed with each flavone in the order Luteolin>Apigenin>Chrysin
In our study, DNMT inhibition by plant flavones seems to be due to the binding of flavones at the catalytic binding pocket of DNMTs. These might be different from catechol-containing polyphenols, such as epigallocatechic-3-gallate, in which the noncompetitive inhibition of DNA methylation catalyzed by DNMTs is majorly due to the higher levels of SAM resulting from the catechol-O-methyltransferase-mediated O-methylation of these compounds.
Together, our study demonstrated the dual potential of flavones in targeting DNMTs and EZH2, which is better than targeting a single silencing enzyme.https://journals.plos.org/plosone/article?id=10.1371/journal.pone.0162956This may imply that even primary POIS could be epigenetic in origin.
DNA methylation typically regulates gene expression by repressing transcription but in some cases DNA methylation can be transcriptionally activating.
These studies provide evidence that early life exposure to endocrine disruptors, and potentially endogenous hormones, do not necessarily elicit immediately detectable changes in DNA methylation or gene expression. In some cases, these responses are not detectable until later in life-puberty or in response to a 'second hit' stimulus such as changes in hormone levels.
It has been proposed that a reawakening of developmental signals drives inappropriate growth in prostate cancer and BPH.https://sci-hub.se/https://www.futuremedicine.com/doi/abs/10.2217/EPI.15.8Recently, interplay between DNA methylation and prostate-associated diseases has been found. The most studied 5-alpha reductase, SRD5A2, has CpG regions, and its expression, which is varied in adulthood, is regulated by Dnmt1. Increased methylation of the SRD5A2 promoter is associated with increasing age in humans and with decreased expression. Thus, DNA methylation could be a personalized medical target for the management of BPH patients resistant to 5-alpha reductase inhibitor therapy.Epithelial-mesenchymal transition (EMT) can lead to prostatic malignancies like
benign prostatic hyperplasia (BPH) or prostate cancer.
During EMT E-cadherin (Cdh1) is decreased.Inflammation is regarded as one of the potential inducers of EMT. Indeed, our results showed that IL-6 and TGF-B1 decreased E-cadherin expression and increased the expression and activity of DNMT1, which in turn induced DNA hypermethylation in the promoter region of Cdh1. Moreover, we found that the combination of the DNMT1 inhibitor 5-Aza and TGF-B1 antibody significantly rescued E-cadherin expression, providing a promising strategy for the treatment of BPH. Cdh1 expression is inappropriately silenced by DNA methylation in breast cancers and prostate cancer. In the present study of BPH, our results showed that the expression of Dnmt1 was higher in BPH cells. It was found that three key downstream mediators in TGF-B signaling (Smad3, Snail, and Slug) were up-regulated in BPH tissue.
Chen et al. showed that TGF-B1 induced EMT through non-canonical PI3K/AKT and MAPK/ERK1/2 signaling pathways. The canonical Smad pathway of TGF-B1 signaling can be enhanced by LPS/TLR4 signaling through the down-regulation of a member of the TGF-B type I receptor family. In prostate cancers, TGF-B1 was identified as activating Erk and therefore Dnmts, which could result in promoter DNA hypermethylation of its own receptors.
Other EMT features, such as N-cadherin, Vimentin and MMP9, should also be fully assessed. Second, the role of other factors, such as IL-8 and TNF-a, should also be investigated in inducing EMT because it has been shown that EMT could be induced by TNF-a through the NF-kB signaling pathway.https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5432940/Imatinib is a potent TNF-a inhibitor that cured some POISers. Suppressing TNF-a downregulates methylation and restores prostatic SRD5A2. However it has to be noted that another study claims imatinib as a DNMT1 inducer, which makes it difficult to make a straight conclusion (see below).
Artificial induction of inflammation in prostate primary epithelial cells leads to hypermethylation of the SRD5A2 promoter and silencing of SRD5A2, whereas inhibition with TNF-a inhibitor reactivates SRD5A2 expression.
We have shown that the SRD5A2 gene contains a promoter with a rich CpG island capable of being methylated. Because epigenetic factors can regulate the expression of genes, we hypothesized that methylation of SRD5A2 leads to reduced SRD5A2 gene expression and protein production during adulthood in benign prostatic tissues. We show that members of the DNA methyltransferase (DNMT) family regulate methylation of SRD5A2. Specifically, we show that DNMT1, and not DNMT3a or DNMT3b, regulates methylation of the SRD5A2 gene promoter.
We found that at baseline, DNMT1 was expressed in BPH-1 cells, however, on exposure to AZA (demethylating compound), there was a significant reduction in DNMT1 levels that was accompanied by increased SRD5A2 expression. These data suggest that DNMT1 plays an important role in silencing SRD5A2 expression.
The results suggest that cytokines including TNF-a and transcriptional factors such as NF-kB (p65) contribute to silencing of SRD5A2 by regulating DNMT1 activity. Therefore, NF-kB/p65 plays a central role not only in increasing the expression of DNMT1, but also in facilitating binding of DNMT1 to the SRD5A2 promoter region, leading to methylation and suppression of the SRD5A2 gene.
By suppressing p65 we found significant down-regulation of IL-6, p-STAT3, and DNMT1, in addition to a significant reduction in DNMT1 activity. However, simultaneous ectopic expression of IL-6 in p65-suppressed cells led to re-expression of DNMT1 protein levels. These results show that NF-kB/p65 regulates DNMT1 binding to the SRD5A2 promoter via an IL-6–dependent pathway.
These results from our patient cohort suggest that increasing age is associated strongly with increased methylation of the SRD5A2 promoter region and reduced SRD5A2 protein expression.
Combined, these results show that increased expression of DMNT1, methylation of the SRD5A2 promoter, and reduced expression of SRD5A2 protein are associated strongly with increased inflammatory mediators with aging.
For example, histone deacetylation, nucleosome occupancy, or gene modifications all can account for other mechanisms that may modulate the expression of SRD5A2.
Although methylation of tumor-suppressor genes and their role in initiation and progression in neoplasia is well studied, the idea that epigenetic changes may be involved in benign disease models is less appreciated. Because the prostate gland is the only solid organ that grows during adulthood, we postulated that epigenetic modifications may play an important role in mediating expression of SRD5A2 in a manner similar to that observed in tumor growth. In this study, we show for the first time that epigenetic modifications of SRD5A2 exist and are regulated by DNMT1.https://www.sciencedirect.com/science/article/pii/S0002944014006907Given the list TGFB downregulation should be beneficial for POISers, though some of them were detrimental for me.
Upregulated transforming growth factor-beta (TGFB) signalling, driving mesenchymal cells to increase their production of ground substance and undergo a transition to a myofibroblast phenotype, is believed to play a pathogenic role in diverse fibrotic disorders, including benign prostatic hyperplasia, scleroderma, pulmonary fibrosis, glomerulosclerosis, tubulointerstial fibrosis, hepatic fibrosis, open angle glaucoma, Peyronie’s disease and the cardiac fibrosis associated with cardiac hypertrophy and heart failure. This issue is now of particular interest, as pulmonary fibrosis is emerging as a not-uncommon long-term complication of COVID-19.
However, the Smad2/3-Smad4 heterodimer quite frequently functions in concert with an AP-1 complex to mediate TGF?-induced transcription. Activation of AP-1 reflects concurrent TGFB-mediated activation of the mitogen-activated protein (MAP) kinases ERK, JNK and p38.
Activated ERK1/2, JNK and p38 MAP kinase can collaborate to boost c-Fos expression and confer a serine phosphorylation on c-Jun which boosts its transactivational activity. As a result, AP-1 activity is markedly induced, and this collaborates with Smad2/3-Smad4 heterodimers to promote TGFB-mediated transcription.
TGFB signalling can be opposed by cGMP, the ligand-bound oestrogen receptor-B (ERB), activation of the nrf2 transcription factor and the Sirt1 deacetylase.
Ligand-bound activated ERB has been found to downregulate TGFB-mediated transcription by a direct interaction with AP-1 complexes that blocks their transactivational activity. This interaction does not involve binding of ERB to oestrogen response elements on DNA, but rather to c-Jun. This may rationalise preclinical and epidemiological evidence that endogenous or therapeutic oestrogen provides protection from cardiac hypertrophy, hepatic fibrosis, glomerulosclerosis and primary open-angle glaucoma (POAG). Pertinently, ERB is expressed in hepatic stellate cells, mesangial cells, cardiac fibroblasts and prostate stroma. Moreover, polymorphisms of the ERB gene (but not that of the ERa gene) have been linked to increased risk for POAG.
In summary, spirulina/PhyCB, phase II inducers such as lipoic acid, ferulic acid or broccoli sprout powder, melatonin, berberine, high-dose biotin, soy isoflavones, taurine and NAC may have potential for downregulating TGFB signalling, and thereby decreasing risk for, or improving clinical control of, a wide range of pro-fibrotic pathologies. Suggested dose schedules for these agents are presented in box 1. With regard to post-COVID-19 syndrome specifically, the antioxidant/anti-inflammatory effects of PhyCB, phase II inducers, melatonin and NAC might address neurological aspects of this syndrome thought likely to reflect chronic inflammation of cerebrovascular endothelial cells and microglia.A more complete list for TGFB downregulation: spirulina/phycocyanin/phycocyanobilin (PhyCB), biliverdin/bilirubin, lipoic acid, ferulic acid, broccoli sprout extracts (sulforaphane), melatonin, NAD+, berberine, metformin, citrulline and/or high-dose folate, sodium nitrate or of nitrate-rich beetroot juice, soy isoflavones, equol, genistein, hydrogen sulfide (H2S), taurine, high-dose biotin, N-acetylcysteine (NAC).
Spirulina also protects E-cadherin.
https://openheart.bmj.com/content/8/1/e001663.fullFulvestrant and decitabine are DNMT inhibitors that increase ERB expression.
Fulvestrant inhibits growth of triple negative breast cancer and synergizes with tamoxifen in ERa positive breast cancer by up-regulation of ERB. Inhibition of DNA methyltransferase (DNMT) increased the levels of ERB and fulvestrant exerted similar potency on DNMT activity as the DNMT inhibitor decitabine. ERB expression may be regulated by methylation of the promoter region of ERB. This process is dependent on DNA methyltransferase (DNMT). Here we report that the pure anti-ERa drug fulvestrant increased ERB expression both at mRNA and protein levels in ERa+/ERB+ as well as in ERa-/ERB+ breast cancers.https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5302959/Imatinib increases EZH2 and DNMT3A. It is likely that imatinib both increases and decreases methylation of certain genes.
Imatinib causes epigenetic alterations of PTEN gene via upregulation of DNA methyltransferases and polycomb group proteins. Histone-methyltransferase enhancer of zeste homolog 2 (EZH2) is essential for DNMTs to bind to the EZH2-target promoters such as MYT1, KCNA1 and cannabinoid receptor 1 (CNR1), and silences expression of these genes. Imatinib-induced DNMT3A formed a complex with EZH2, which facilitated their binding to the PTEN promoter and induced DNA hypermethylation of this region, leading to downregulation of PTEN. Imatinib increased levels of both EZH2 and DNMT3A in leukemia cells, which probably caused global epigenetic aberrations and downregulated the expression of important genes involved in regulation of cell growth, apoptosis and drug metabolism, which could relate to acquisition of drug resistance.
The drug resistance against tyrosine kinase inhibitors could be overcome by treatment with an anti-epigenetic agent histone deacetylase inhibitor, highlighting a potential therapeutic strategy. In fact, we previously showed that histone deacetylase inhibitor successfully overcame imatinib resistance in EOL-1R cells in association with restoration of phosphatase and tensin homolog deleted on chromosome 10 (PTEN) expression.https://www.nature.com/articles/bcj201133.pdf?origin=ppubSome POISer had success with a high-fat diet (HFD).
We showed a lower expression of Dnmt1 in colon with HFD with no changes due to vitamin E supplementation, although in CD animals an increase with supplementation was noted.
Vitamin E reduced DNA damage and showed organ-specific effects on MLH1 and Dnmt1 gene expression and methylation. The repair protein MutL homolog 1 (MLH1) is part of the DNA mismatch repair (MMR) system. Microsatellite instability (MSI) is often associated with promoter hypermethylation, resulting in inactivation of MLH1.
In mice with diet induced obesity (DIO), the binding of HDACs is increased at the leptin promoter. In another DIO mouse model, Dnmt1 expression and enzymatic activity were elevated in adipocytes, leading to promoter hypermethylation and following decreased adiponectin expression.https://www.mdpi.com/2072-6643/9/6/607/htmOur study suggests that curcumin may represent an effective antioxidant compound against diabetic retinopathy (DR), via restoring oxidative stress and DNMT functions, though further studies are recommended.
There are different types of repetitive sequences scattered throughout the genome (e.g., satellite repeat, short interspersed nuclear element, and long interspersed nuclear element-1 (LINE-1)). LINE-1 sequences, accounting for about 18% of human genome, are widely used as a surrogate marker of global DNA methylation.
Consistently, the high glucose-induced effect on DNMT expression was evident after 48 hours from the insult, with the upregulation of DNMT1. These changes persisted even when the glucose level is normalized, indicating that DNA methylation is probably involved in the metabolic memory of DR.
The early effect results in decreasing DNMT activity, accompanied by the highest ROS production, while long-term oxidative stress increases DNMT activity and DNMT1 expression.https://www.hindawi.com/journals/omcl/2018/5407482/Genistein (2–20 umol/l) was found to inhibit DNMT, reverse DNA hypermethylation and reactivate RARB, p16, and O6-methylguanine methyltransferase (MGMT) in esophageal squamous carcinoma and prostate and mammary cancer cells in vitro. In theory, prevention or reversal of hypermethylation-induced inactivation of key tumor suppressor or receptor genes by DNMT inhibitors such as genistein and daidzein could be an effective approach for cancer prevention, but little is known about the methylation effects of these compounds in humans.https://sci-hub.se/https://www.tandfonline.com/doi/abs/10.1080/01635580802404196Vitamin D is popular amongst POISers and drug resistance was an issue for some.
These data suggest that 1,25(OH)2D3 treatment reduces expression and activity of DNMT1 and DNMT3B and may lead to reduced DNA methylation.
Our results suggest that long-term 1,25(OH)2D3 treatment induces hypomethylation and overexpression of specific genes that may contribute to vitamin D resistance.
Overall, these hypomethylated/upregulated genes are involved in long-term vitamin D treatment-induced upregulation of the mTOR signaling axis. In a recent animal study, suppressed mTOR signaling is observed in the rat model of chronic vitamin D deficiency, and restoring vitamin D in rats reverses the effect. The mTOR pathway inhibitor metformin has been reported to increase the growth-inhibitory effects of vitamin D. In addition, the combination of vitamin D and metformin synergistically prevents colon carcinogenesis in both rat and mouse colon neoplasia models. Since long-term 1,25(OH)2D3 treatment is confirmed to induce hypomethylation and overexpression of genes that lead to overactivation of the mTOR pathway, future combinations of mTOR inhibitors with vitamin D therapy may be useful in preventing vitamin D resistance.https://journals.physiology.org/doi/full/10.1152/ajpcell.00522.2019Curcumin's ability to inhibit HAT could be one reason why it doesn't work for me.
Several vitamin Bs, especially folic acid and vitamin B12, may have roles in DNA synthesis, repair, and methylation in which methylenetetrahydrofolate reductase (MTHFR) plays a critical key of catalyzing the synthesis of 5-methyltetrahydrofolate, therefore, regulating the utility of diet folate in DNA metabolism pathways.
In addition to the classic dietary HDAC inhibitor, butyric acid, more recently there are a number of dietary HDAC inhibitors being discovered including diallyl disulfite and sulforaphane. Diallyl disulfite is a garlic compound and sulforaphane is a major isothiocyanate found in broccoli and cauliflowers.
Interestingly instead of themselves, their metabolites, S-allylmercaptocysteine, and sulforaphane-cysteine respectively have been identified as the true HDAC inhibitors.
On the other hand, it was reported that a dietary polyphenol, curcumin, can induce histone hypoacetylation in vitro and in vivo by inhibiting histone acetylase (HAT) instead of HDAC. The reactive oxygen species generated by curcumin may be involved in the inactivation of HAT. Folate, vitamin B12, selenium may have certain critical roles in DNA methyl metabolism pathways. Flavonoids (dietary catechols) like caffeic acid, chlorogenic acid, quercetin, myricetin, tea catechins and others seem to show an indirect mechanism to inhibit DNMT activity to achieve reactivation of silenced genes in cancer cells. COMT in mammalian cells can catalyze SAM (also the substrate of DNMT) mediated O-methylation of the above dietary catechols and the other product, SAH (a potent inhibitor for DNMT), thus, by competing the same substrate (i.e., SAM) and producing a potent inhibitor, SAH, the DNMT activities can be effectively inhibited by the above phytochemicals.https://sci-hub.se/https://www.ingentaconnect.com/content/ben/ccdt/2007/00000007/00000007/art00010A vitamin A derivate used in acne therapy (isotretinoin) has been inconsistently associated with the onset of IBD. However, what needs to be considered is the previous treatment of acne patients with antibiotics that are also associated with the development of IBD, thus representing a crucial confounding factor. Here, we studied whether doxycycline (acne therapy), metronidazole (IBD therapy) or isotretinoin are able to induce alterations in DNA methylation and microRNA expression patterns in murine colonic intestinal epithelial cells (IECs).
As for changes in DNA methylation, we found isotretinoin to have strong demethylating effects, while antibiotic treatment had only a moderate impact.
Regarding microRNA and mRNA expression, isotretinoin and doxycycline, but not metronidazole, potentially induce long-term changes in microRNA/mRNA expression profiles towards the down-regulation of immune responses.
In contrast, isotretinoin had a strong impact on DNA methylation with 234 hypomethylated targets compared to the vehicle. Previous treatment with isotretinoin resulted in eight hypermethylated targets, indicating a resolution of the direct effect on demethylation, and thereby pointing to a short-term effect of isotretinoin on DNA methylation. In total, 308 mRNAs were up-regulated and 136 mRNAs down-regulated after isotretinoin-washout, while treatment with metronidazole and doxycycline up-regulated the expression of 23 and 59 mRNAs, respectively, and down-regulated the expression of 59 and 91 mRNAs, respectively. These results point to possible long-term changes on mRNA expression levels after isotretinoin treatment in murine colonic IECs.
Treatment with the vitamin A derivate isotretinoin led to the demethylation of targets that were involved in IL-12 signaling and developmental processes, including the hypomethylated target IL-12rb1 and the corresponding up-regulated IL-12-dependent pathway in the transcriptome analysis directly after isotretinoin treatment in IECs. Although epigenetic marks such as DNA methylation are long-lasting and inheritable modifications, isotretinoin-induced demethylation was resolved after the washout phase of 4 weeks, indicating short-term effects on IL-12rb1. Furthermore, isotretinoin did not affect IBD-associated microRNAs in IECs neither directly after treatment cessation nor after the washout phase.https://www.mdpi.com/2075-4655/1/3/24/htmHDAC1 is overexpressed in prostate, gastric, colon, and breast cancers, and HDAC2 is overexpressed in colorectal, cervical, and gastric cancers.
Direct effects may be caused by drugs that affect chromatin architecture or DNA methylation, whereas indirect effects may be caused by drugs that affect transcription. For example, hydralazine directly affects the epigenome by inhibiting DNA methylation, and isotretinoin indirectly affects the epigenome by altering transcription factor activity. Drugs that indirectly affect the epigenome initially influence signaling pathways, thereby altering transcription factor activity at gene promoters and subsequently altering expression of receptors, signaling molecules, and other proteins. One study postulated that cells will ultimately adopt more permanent modifications to DNA methylation and chromatin structure, leading to enduring epigenetic changes. Thus these effects could persist after the drug is discontinued.https://sci-hub.se/https://accpjournals.onlinelibrary.wiley.com/doi/abs/10.1002/phar.1408In the meanwhile I tested a Magnolia bark supplement and it provided a moderate benefit for me.
Honokiol is a phytochemical that can be found in Magnolia species. Honokiol could prevent tumorigenesis.
Honokiol could significantly inhibit DNMT activity and reduced Dnmt1, Dnmt3a and Dnmt3b proteins expression. Honokiol could also reactivate and restore the levels of TET proteins suppressed by UVB radiation.
Indomethacin can also inhibit DNA methylation. Honokiol application induced persisting effects.
Honokiol application also inhibited UVB-induced DNA hypermethylation and its elevation of the levels of TET enzyme, which is responsible for DNA demethylation in UVB-exposed skin.
Collectively, these data suggest that PGE2 promotes UVB-induced immunosuppression and that the PGE2 may act to suppress the immune reactivity by promoting DNA methylation in UVB-exposed skin.
It has been shown that the ten eleven translocation (TET) enzymes that catalyze demethylation of 5-methyl cytosine (5-mC) and promote locus-specific reversal of DNA hypermethylation.
Furthermore, honokiol inhibits the transcription regulators of Dnmt activity, Sp1 and Sp3, which have been implicated in DNA demethylation. In addition to blocking the addition of extra methyl groups to the 5th position of cytosine through inhibiting Dnmt activity, honokiol treatment also promotes DNA demethylation of existing DNA hypermethylation through activation of the TET enzyme and expression of TET proteins. Active DNA demethylation by TET proteins has been shown to play critical roles in T cell functions and particularly cytokine expression. This is consistent with the literature indicating that epigenetic modifications in general, and especially aberrant DNA methylation, play important role in the regulation of cytokines in malignancies.https://www.nature.com/articles/s41598-017-01774-5T-cells are implicated in POIS.
Perhaps consistent with such antagonism between DNMT1 and TET in Treg cells, knockdown of DNMT1 activity induces the expression of Foxp3 in conventional CD4 T cells whereas loss of TET protein activity leads to unstable Foxp3 expression.
Both DNMT1 and TET enzymatic activities are highly sensitive to the metabolic state of T cells. Unlike T effector (Teff) cells that rely heavily on aerobic glycolysis for energy generation, stable Foxp3 + Treg cells generate little lactate in the presence of glucose and instead make use of lipid and glucose oxidative phosphorylation (OXPHOS) and mitochondrial electron transport for ATP synthesis.
One long-standing question in the investigation of anergy as a peripheral immune tolerance mechanism has been its purpose. Why should the immune system actively promote the survival of potentially dangerous self-reactive T cells when mechanisms exist to delete such cells from the repertoire? One attractive hypothesis is that anergy reversal can at times be protective—either to facilitate aggressive immunity against particular tissue-specific self-antigens during intracellular infection or cancer or to augment antigen-specific suppression in the face of immunopathology.
Therefore, we now hypothesize that partial demethylation of the Foxp3 CNS2 region is a key feature of anergic Treg progenitor cells.
Therefore, DNMT1 activity and Foxp3 CNS2 maintenance methylation may destabilize Foxp3 expression in the setting of T-cell lymphopenia.
Thus, we hypothesize that high DNMT1 and SAM-e levels, chromosomal replication, and Foxp3 CNS2 remethylation promote the differentiation of dangerous conventional CD4 Teff cells following the reversal of anergy. This is particularly true when an anergic T cell has accumulated only a modest number of hydroxymethylated and demethylated CpG nucleotides at the CNS2 locus.https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6305231/The above information could mean that taking a combination of DNMT1 and HDAC inhibitors for a prolonged time may actually cure POIS for at least a group of us. Some case reports show that Relora (honokiol) was capable of curing at least one POISer. Medicinal mushrooms (beta-glucan) could also result in sustained symptom amelioration. Thus a combination like beta-glucan (medicinal mushrooms, barley, oat straw), honokiol (Magnolia/Relora), resveratrol, chrysin (passion flower/noni), etc. can potentially cause permanent effects on disease severity or complete resolution over time, though primary cases may not benefit as much as secondary ones. Other POIS cases could be caused by hypomethylated genes and such a therapy may be detrimental for them, although more information would be required to properly differentiate POIS types.
Some other opportunities for treatment of this type could be an increase of E-cadherin, inhibition of TGFbeta, increase of TET, inhibition of TGF-a and IL6, decreasing EZH2 and many more options can be derived from the above. Needless to say this is not only a treatment model for POIS, but a number of cancers, neurodegenerative diseases, viral infections and a number of other ailments. Researchers have already realized this, it just hasn't become common knowledge so far.