Butyrate - Health benefits, anti-inflammatory and key for the microbiota-gut-brain axis

Gaby

SuperModerator
Moderator
FOTCM Member
It is thought that fecal transplants work because they restore butyrate levels in the gut. We have information about butyrate all over the forum, but here's a dedicated thread to raise up awareness into this important short-chain fatty acid.

Butyrate is produced in the small intestines and is also known as butyrate acid or butanoic acid. Good bacteria in the intestines produce butyrate when they ferment dietary fiber.

From wiki Butyric acid - Wikipedia :

Butyrate is essential to host immune homeostasis.[36] Although the role and importance of butyrate in the gut is not fully understood, many researchers argue that a depletion of butyrate-producing bacteria in patients with several vasculitic conditions is essential to the pathogenesis of these disorders. A depletion of butyrate in the gut is typically caused by an absence or depletion of butyrate-producing-bacteria (BPB). This depletion in BPB leads to microbial dysbiosis. This is characterized by an overall low biodiversity and a depletion of key butyrate-producing members. Butyrate is an essential microbial metabolite with a vital role as a modulator of proper immune function in the host. It has been shown that children lacking in BPB are more susceptible to allergic disease[46] and Type 1 Diabetes.[47] Butyrate is also reduced in a diet low in fiber which can induce inflammation and have other adverse affects insofar as these short-chain fatty acids activate PPAR-γ.

Butyrate exerts a key role for the maintenance of immune homeostasis both locally (in the gut) and systemically (via circulating butyrate). It has been shown to promote the differentiation of regulatory T cells. In particular, circulating butyrate prompts the generation of extrathymic regulatory T cells. The low-levels of butyrate in human subjects could favor reduced regulatory T cell-mediated control, thus promoting a powerful immuno-pathological T-cell response.[49] On the other hand, gut butyrate has been reported to inhibit local pro-inflammatory cytokines. The absence or depletion of these BPB in the gut could therefore be a possible aide in the overly-active inflammatory response. Butyrate in the gut also protects the integrity of the intestinal epithelial barrier. Decreased butyrate levels therefore lead to a damaged or dysfunctional intestinal epithelial barrier.
Butyrate has been shown to be a critical mediator of the colonic inflammatory response. It is responsible for about 70% of energy from the colonocytes, being a critical SCFA in colon homeostasis.[65] Butyrate possesses both preventive and therapeutic potential to counteract inflammation-mediated ulcerative colitis (UC) and colorectal cancer.[66] It produces different effects in healthy and cancerous cells: this is known as the "butyrate paradox". In particular, butyrate inhibits colonic tumor cells and stimulates proliferation of healthy colonic epithelial cells.

In summary, the production of volatile fatty acids such as butyrate from fermentable fibers may contribute to the role of dietary fiber in colon cancer. Short-chain fatty acids, which include butyric acid, are produced by beneficial colonic bacteria (probiotics) that feed on, or ferment prebiotics, which are plant products that contain dietary fiber. These short-chain fatty acids benefit the colonocytes by increasing energy production, and may protect against colon cancer by inhibiting cell proliferation.

Owing to the importance of butyrate as an inflammatory regulator and immune system contributor, butyrate depletions could be a key factor influencing the pathogenesis of many vasculitic conditions. It is thus essential to maintain healthy levels of butyrate in the gut. Fecal microbiota transplants (to restore BPB and symbiosis in the gut) could be effective by replenishing butyrate levels. In this treatment, a healthy individual donates their stool to be transplanted into an individual with dysbiosis. A less-invasive treatment option is the administration of butyrate—as oral supplements or enemas—which has been shown to be very effective in terminating symptoms of inflammation with minimal-to-no side-effects. In a study where patients with ulcerative colitis were treated with butyrate enemas, inflammation decreased significantly, and bleeding ceased completely after butyrate provision.
 
From Potential beneficial effects of butyrate in intestinal and extraintestinal diseases

The SCFA butyrate, a main end product of microbial fermentation of dietary fibers in the human intestine, plays an important role in the maintenance of intestinal homeostasis and overall health status. The effects exerted by butyrate are multiple and involve several distinct mechanisms of action. Its well-known epigenetic mechanism, through the inhibition of HDACs, results in the regulation of gene expression and in the control of cell fate. At the intestinal level, butyrate exerts multiple effects such as the prevention and inhibition of colonic carcinogenesis, the improvement of inflammation, oxidative status, epithelial defense barrier, and the modulation of visceral sensitivity and intestinal motility. At the extraintestinal level, potential fields of application for butyrate seem to be the treatment of sickle cell disease, β-thalassemia, cystic fibrosis, urea cycle enzyme deficiency, X-linked adrenoleukodystrophy, hypercholesterolemia, obesity, insulin resistance, and ischemic stroke. In conclusion, a growing number of studies have revealed new mechanisms and effects of butyrate with a wide range of potential clinical applications from the intestinal tract to peripheral tissues.

Canani RB, Costanzo MD, Leone L, Pedata M, Meli R, Calignano A. Potential beneficial effects of butyrate in intestinal and extraintestinal diseases. World J Gastroenterol 2011; 17(12): 1519-1528 [PMID: 21472114 DOI: 10.3748/wjg.v17.i12.1519]
 

Several lines of evidence suggest that brain function and behaviour are influenced by microbial metabolites. Key products of the microbiota are short-chain fatty acids (SCFAs), including butyric acid. Butyrate is a functionally versatile molecule that is produced in the mammalian gut by fermentation of dietary fibre and is enriched in butter and other dairy products. Butyrate along with other fermentation-derived SCFAs (e.g. acetate, propionate) and the structurally related ketone bodies (e.g. acetoacetate and d-β-hydroxybutyrate) show promising effects in various diseases including obesity, diabetes, inflammatory (bowel) diseases, and colorectal cancer as well as neurological disorders. Indeed, it is clear that host energy metabolism and immune functions critically depend on butyrate as a potent regulator, highlighting butyrate as a key mediator of host-microbe crosstalk. In addition to specific receptors (GPR43/FFAR2; GPR41/FFAR3; GPR109a/HCAR2) and transporters (MCT1/SLC16A1; SMCT1/SLC5A8), its effects are mediated by utilisation as an energy source via the β-oxidation pathway and as an inhibitor of histone deacetylases (HDACs), promoting histone acetylation and stimulation of gene expression in host cells. The latter has also led to the use of butyrate as an experimental drug in models for neurological disorders ranging from depression to neurodegenerative diseases and cognitive impairment.
Here we provide a critical review of the literature on butyrate and its effects on multiple aspects of host physiology with a focus on brain function and behaviour. We find fundamental differences in natural butyrate at physiological concentrations and its use as a neuropharmacological agent at rather high, supraphysiological doses in brain research. Finally, we hypothesise that butyrate and other volatile SCFAs produced by microbes may be involved in regulating the impact of the microbiome on behaviour including social communication.
 

Attachments

  • j.neuint.2016.06.011.pdf
    2.7 MB · Views: 7

Emerging evidence suggests that the intestinal microbiota is a source of sleep-promoting signals. Bacterial metabolites and components of the bacterial cell wall are likely to provide important links between the intestinal commensal flora and sleep-generating mechanisms in the brain. Butyrate is a short-chain fatty acid produced by the intestinal bacteria by the fermentation of nondigestible polysaccharides. We tested the hypothesis that butyrate may serve as a bacterial-derived sleep-promoting signal. Oral gavage administration of tributyrin, a butyrate pro-drug, elicited an almost 50% increase in non-rapid-eye movement sleep (NREMS) in mice for 4 hours after the treatment. Similarly, intraportal injection of butyrate led to prompt and robust increases in NREMS in rats. In the first 6 hours after the butyrate injection, NREMS increased by 70%. Both the oral and intraportal administration of butyrate led to a significant drop in body temperature. Systemic subcutaneous or intraperitoneal injection of butyrate did not have any significant effect on sleep or body temperature. The results suggest that the sleep-inducing effects of butyrate are mediated by a sensory mechanism located in the liver and/or in the portal vein wall. Hepatoportal butyrate-sensitive mechanisms may play a role in sleep modulation by the intestinal microbiota.
 

Butyric acid or butyrate can act primarily as an anti-inflammatory molecule and various studies have reported its role in mitigating hyperinflammation via several mechanisms 2527. For the past several years, our group has worked on the role of proinflammatory regulators in the pathogenesis of various inflammatory disorders and identified that the role of histone deacetylase (HDAC) inhibitor in activating anti-inflammatory molecules. Further, this leads to the simultaneous down regulation of proinflammatory membrane receptors, downstream signalling molecules and respective cytokines, resulting in inflammatory homeostasis. Our in vitro preliminary experiments using various cell lines have revealed that the molecular mechanism of butyrate in neutralising inflammatory devastation, induction of anti-inflammatory molecular expression and its translocation to the site of action, is almost similar to dexamethasone 28. Consequently, we hypothesise if the SCFA, butyric acid, a HDAC inhibitor, which is synthesized by the gut microbiota, could have strong anti-inflammatory functions with anti-fibrotic properties. Therefore, this article reviews the anti-inflammatory properties of butyric acid or butyrate and its associated molecular pathways involved in controlling the cytokine storm and hyperinflammation associated with SARS-CoV-2 infection. Based on the various positive reports, we presume that butyric acid possesses potent anti-inflammatory activity, which suggests it as an alternative to dexamethasone for the preventive management of primary and secondary complications related to CoVID-19.

Nutrients in mitigating the Covid-19 pathogenesis​

Nutrition and nutrients play a vital role in enhancing immune response along with reduction of inflammation and oxidative stress 4446. Better nutritional status of CoVID-19 patients is associated with less adverse outcomes 18, 4752. Vitamin D is involved in reducing respiratory infections, such as influenza, and a reduced plasma 25-hydroxyvitamin D (25(OH)D) concentration in SARS-CoV-2 patients has been observed 53. Moreover, people with vitamin D deficiency are at higher risk of getting infected with SARS-CoV-2 54, 55. Co-supplementation of vitamin D along with glutathione precursor L-cysteine significantly increases serum 25(OH)D levels and augments vitamin D regulatory gene expression, which in turn reduces the oxidative stress and inflammatory responses in CoVID-19 patients 56. Vitamin D supplementation in SARS-CoV-2 infected patients attenuates the production of proinflammatory cytokines like Interferon (IFN)-γ, IL-6, IL-2 and TNF-α by inhibiting NF-κB and other pathways 5759. CoVID-19 associated inflammatory signalling pathways including NF-κB, Mitogen-Activated Protein Kinase (MAPK) and phosphatidylinositol 3-kinase/ protein kinase B (PI3K/AKT) and innate immune response pathways, such as Toll-like signalling and NOD-like signalling modulation and regulation can be mediated by the combination of curcumin, vitamin C, and glycyrrhizic acid 60. Vitamin C has been known to improve the immune condition by enhancing differentiation and proliferation of B- and T-cells, but severe vitamin C deficiency is associated with pneumonia and respiratory tract infections 61. Intravenous administration of vitamin C can significantly decrease IL-6 levels 62, 63. Glycyrrhizic acid and curcumin exhibits anti-viral, anti-inflammation, anti-cancer, and immune system benefits 60. The combination of vitamin D/magnesium/vitamin B12 significantly reduced the subsequent need for oxygen therapy and/or intensive care support in older CoVID-19 patients 57. Vitamin B12 is crucial in maintaining the healthy gut microbiome which plays a vital role in immune responses 57. Fat soluble vitamin E acts as an antioxidant that scavenges Reactive Oxygen species (ROS) and inhibits devastating effects of hyperinflammation 64. Moreover, the supplementation of vitamin E stimulates T cell function and confers protection against upper respiratory infections 65.
Selenium is one of the key micronutrients known to positively impact CoVID-19 patient recovery 66, 67. Selenium status regulates the expression of glutathione peroxidase 1 (GPX1), a cytosolic selenoenzyme known for its antioxidative properties. The antioxidant enzyme GPX1 mitigates the production of ROS and further leading to mutations in the viral genome 68. In addition, attenuating ROS also helps in the inhibition of proinflammatory NF-κB activation and further nuclear translocation 69. Severe endothelial injury and widespread pulmonary micro thromboses are accompanied with platelet activation and aggregation in patients with severe CoVID-19 manifestations. The synthetic Rupatadine (histamine1 receptor antagonist) and natural flavonoids with anti-inflammatory properties are known to inhibit the platelet activating factor 70. Elderly individuals with deficiency of nutrients, such as vitamin C, vitamin D, calcium, folate, and zinc are prone to increase severity of SARS-CoV-2 infection 71. Folic acid may inhibit furin protease and inactivates chymotrypsin-like protease (3CL pro) 72. Zinc (Zn 2+) deficiency contributes to impaired cell mediated immune response and increased susceptibility to various infections. However, increased intracellular levels of Zn 2+ disrupt viral RNA replication including SARS-CoV-2, where Zn 2+ inhibits RNA (Ribonucleic acid) dependent RNA polymerase (RdRp) elongation and template binding 73. Among CoVID-19 patients, iron deficiency is strongly associated with increased inflammation and longer stay in hospitals 74.
Health beneficial compounds, including minerals, antioxidants, phytochemicals, vitamins, and minerals present in fruits and vegetables, can exert antioxidative, anti-inflammatory and antiviral effects during various non-infectious and infectious disease 71. Alliin, an S-allyl cysteine sulfoxide compound present in garlic has shown to have inhibitory action on 3CL pro, a protease that plays a vital role in SARS-CoV-2 replication 75. Salvianolic acid A and curcumin have the potential to bind to 3CL pro with greater affinity 76. Resveratrol acts as an anti-inflammatory molecule that inhibits the NFκB pathway and thereby reduces circulatory cytokines, such as IL-6 and TNF-α levels, which are observed in severe SARS-CoV-2 infection 77. Sea cucumber ( Stichopus japonicus) derived sulphated polysaccharide showed significant anti-viral activity against SARS-CoV-2 infection 78. Omega-3 polyunsaturated fatty acids, including eicosapentaenoic acid and docosahexaenoic acid have been shown to exhibit anti-inflammatory effects by downregulation of the NF-κB pathway 71, 79, 80. Free fatty acids such as oleic acid, arachidonic acid and linoleic acid have shown antiviral activity at micromolar concentrations 81. Dietary fibre intake alters the intestinal microflora and enhances relative proportion of SCFAs, which exhibit anti-inflammatory properties through fatty acid receptors like G-protein-coupled receptor (GPCR) 41 and 43 8284.
Go to:

Probiotics: suppressors of respiratory tract infections and inflammation​

Probiotics are living microorganisms that provide health benefits to the host upon administration at appropriate doses 85. Probiotics exert a wide range of beneficial effects such as host microbiome balancing, stimulation of immune system, enhancement of intestinal barrier function or inhibiting pathogens by direct interactions 40, 46, 86, 87 ( Table 1). Several microorganisms belonging to the family of Enterococcus species ( E. fecalis, E. faecium), Bifidobacterium species ( B. bifidum, B. longum, B. lactis), Lactobacillus species ( L. acidophilus, L. casei, L. rhamnosus), and Saccharomyces ( S. boulardii, S. cerevisiae) are considered as probiotics 40. Probiotic supplementation causes significant reduction in the incidence of oral and respiratory tract infections 88, 89. Dietary supplementation of cow’s milk and fermented rice with L. paracasei CBA L74 helps in prevention of common infectious disease including upper respiratory tract infections in children 90. Daily intake of fermented milk containing probiotic L. casei strain ‘Shirota’ has been shown to reduce the incidence and duration of respiratory tract infections in healthy middle aged office workers and young children via modulation of the immune system 91, 92. Daily ingestion of the probiotic L. paracasei ST11 can reduce the degree of virus replication and dissemination thereby attenuating lung inflammation and subsequent death in mice infected with vaccinia virus 95. L. gasseri SBT2055 exhibits antiviral activity against human respiratory syncytial virus (RSV) by silencing SWI2/SNF2-related cAMP Response Element-Binding Protein (CREB)-binding protein activator protein, which is involved in RSV replication. L. gasseri SBT2055 reduced the expression of proinflammatory cytokines in lungs upon RSV infection 96. CC chemokine receptor 2 acts as a receptor for monocyte chemoattractant protein-1 (MCP-1), which induces increased lung inflammation and subsequently decreases survival associated with influenza virus infection. Prophylactic oral administration of heat-killed E. faecalis can protect mice from influenza virus infection and subsequent lung inflammation by modulation of MCP-1 production. Alternatively, lipoteichoic acid of E. faecalis binds to toll like receptor 2 and exerts antiviral and anti-inflammatory activity during influenza infection 97. Oral administration of probiotics L. paracasei, L. gasseri, and B. longum improved immune response and reduced mortality in influenza infected mice 105 by reducing the inflammation and oxidative stress associated with it 106, 107.
Probiotics, in combination with enteral nutrition, given to post-operative gastric cancer patients aids in increased production of antibodies and reduction of inflammatory cytokines 108. Oral administration of L. plantarum ameliorates intestinal inflammation and lipid metabolism disorders by modulating gut microbiota in turn producing more SCFAs in high-fat diet induced obese mice 100. This disrupted enterohepatic immunoregulation, which can be ameliorated by intervention of Clostridium butyricum B1 via its metabolite butyric acid 99. Probiotic mixture of Lactobacillus and Bifidobacterium prevents the non-alcoholic fatty liver disease by suppressing systemic adiposity and inflammation through butyric acid and its receptor GPR109A 98. Treatment with probiotic strain L. acidophilus DDS-1 upsurges the abundance of beneficial bacteria such as Lactobacillus spp and Akkermansia spp and also the levels of butyrate, while downregulating the production of inflammatory cytokines IL-6, IL-1β, IL-1α, MCP-1, Macrophage Inflammatory Protein (MIP)-1α, MIP-1β, IL-12 and IFN-γ in aging mice 101. L. paracasei KW3110 suppresses hyperinflammation via activation of M2 macrophages and exhibit anti-inflammatory effects via suppression of IL-β production and caspase 1 activation by promoting IL-10 production 103. Probiotic complex of L. acidophilus, L. casei, L. fermentum, L. paracasei, Streptococcus thermophilus, Bifidobacterium longum, B. bifidum, B. breve, L. rhamnosus, L. plantarum, L. helveticus, and L. salivarius in combination with zinc and coenzyme Q10 can improve autoimmune arthritis via downregulation of proinflammatory cytokines including IL-6, IL-17 and TNF-α and inhibition of T-helper cell 17 (Th17) cell differentiation 109111. Oral administration of B. infantis suppresses allergic inflammation in lungs by significantly reducing serum levels of Immunoglobulin (Ig)E, IgG1, IL-4 and IL-13 102 . Daily administration of L. plantarum DR7 for 12 weeks can prevent development of upper respiratory tract infections among young adults through various mechanisms including inhibition of respiratory infection causing bacteria such as Staphylococcus aureus, Streptococcus pneumoniae, Streptococcus pyogenes and Streptococcus mutans, stimulation of proinflammatory cytokine production such as IL-10 and IL-4, and enhancement of antioxidant potential of RBC membrane 93. Significant reduction in the number of Bifidobacteria and Lactobacilli along with increased number of Escherichia coli is observed in the gut of children with recurrent respiratory tract infections. Oral probiotic supplement containing Bifidobacterium infantis, L. acidophilus, E. faecalis and Bacillus cereus restored the intestinal flora along with reduction in incidence of respiratory tract infections and use of antibiotics 23.
Intestinal microflora widely affects host health and alterations in the gut microbiome is correlated with several disease including respiratory disease 130. Commensal gut microbiome and its metabolites can modulate host immunity and can also impact on pro inflammatory and immune-regulatory response 131. Increased production of microbiome metabolite SCFAs may improve health condition 132. Depletion of SCFA production makes mice more susceptible for allergic lung inflammation. Biological effects exerted by SCFAs is dependent mainly on two mechanisms: SCFA mediated (i) activation of GPCRs and (ii) inhibition of HDAC. SCFAs, via HDAC inhibition, positively impacts the functions and numbers of T-helper 1 cells, T-regulatory cells, and Th17 effector cells resulting in reduced inflammatory response in airway diseases 130. The short chain fatty acid, butyrate or butyric acid is produced in the colon by anaerobic bacteria such as Roseburia intestinalis, Faecalibacterium prausnitzii, Clostridium butyricum, Megasphaera elsdenii, Mitsuokella multiacida, Eubacterium spp., Fusobacterium spp., Butyrivibrio spp. and Eubacterium hallii 133. Butyrate concentration in the colon can reach from 10 to 20 mM and serves as major source of energy for colonocytes. Sodium butyrate supplementation enhances the abundance of beneficial bacteria such as Coprococcus, Lachnospiraceae, Ruminococcus, Bifidobacteriaceae and Actinobacteria improving intestinal barrier integrity in obese mice 134.

Primarily, butyric acid exhibits anti-inflammatory and tissue protective function in the large intestine 135. Butyric acid is a potential inhibitor of pro-inflammatory molecule NF-κB 135137 ( Figure 1). Tight junction protein expression in intestinal epithelial cells is also influenced by butyrate mediated regulation 138. Butyrate treatment on epithelial colon cells significantly downregulated the proinflammatory molecules including Toll-like receptor (TLR)2, TLR4, IL-6, IL-12A, IL-1β, IL-18, TNF, MAPK13, MAPK10, MAPK3, AKT1, AKT2, AKT3, NF-κB1A, NF-κB1, CXCL1, CXCL2, CXCL3, CXCL6, CXCL8, Chemokine ligands (CCL)2, Serpin peptidase inhibitor, clade A (alpha-1 antiproteinase, antitrypsin), member 1 (SERPINA1), SERPINA2, Colony Stimulating Factor (CSF) 3, Intercellular Adhesion Molecule 1 (ICAM1), Vascular Endothelial Growth Factor A (VEGFA), Major Vault Protein (MVP), Cathelicidin Antimicrobial Peptide (CAMP) and insulin-like growth factor binding protein (IGFBP)3, along with inhibition of proinflammatory pathways, including (i) triggering receptor expressed on myeloid cells (TREM-1) signalling, (ii) production of nitric oxide (NO) and ROS, (iii) high-mobility group box-1 (HMGB1) signalling, (iv) IL-6 signalling, and (v) acute phase response signalling 25. Pre-treatment with butyric acid can attenuate heart depression along with reduction in inflammation and oxidative stress associated with septic shock in mice 139. Acute lung injury along with ARDS characterized by excessive inflammation can be induced by various factors such as endotoxins, infections, hypoxia and complement activation. Lipopolysaccharide (LPS) induced acute lung injury (ALI) and inflammation can be attenuated by 4-phenyl butyric acid (4-PBA), a derivative of butyric acid and also by sodium butyrate.
Prophylactic treatment of sodium butyrate significantly reduces myeloperoxidase activity and inflammatory cell infiltration into lungs which is correlated with the inhibition of proinflammatory cytokine, HMGB1 expression and NFκB 26. The TLR 4/NF-κB pathway involved in the LPS is targeted by sodium butyrate, which attenuates the LPS induced lung injury 27. Hyaluronan ester with butyric acid treatment induces apoptosis in mesangial cells after exposure to oxidative stress and thereby reducing cell proliferation via p38 MAPK pathway 141. N-(1-carbamoyl-2-phenyl-ethyl) butyramide (FBA), a butyrate releasing compound, confers protection to mice from colitis induced by dextran sodium sulphate by suppressing neutrophils recruitment and subsequent release of pro-inflammatory molecules mediated by HDAC-9/NF-κB inhibition and peroxisome proliferator-activated receptor gamma (PPAR-γ) upregulation 142. Butyrate inhibits IL-13 and IL-15 production by Type 2 innate lymphoid cells. Butyrate downregulates various RNA binding proteins and thereby post transcriptionally downregulating the expression of inflammatory genes 143. Sodium butyrate attenuates AngII induced hypertension, cardiac hypertrophy, cardiac fibrosis, and inflammation by inhibiting Cyclooxygenase-2 (COX2)/ Prostaglandin E2 (PGE2) pathway in a HDAC5/ HDAC6 dependent manner 144. Butyrate reduces AngII induced endothelial dysfunction 145. Sodium butyrate attenuates lung inflammation by promoting forkhead box P3 (FOXP3) expression and suppression of IL-9 expression. Butyrate also reduces the infiltration of proinflammatory Th9 cells and eosinophils into lungs 146. Mice treated with butyrate exhibited a significant reduction of inflammatory infiltrates in the airways, tissue, and vascular disruption, and subsequently less haemorrhaging in the lungs induced by influenza infection 82. HDAC inhibitor sodium butyrate can suppress ACE2 expression in gut epithelial cells which can help in reducing gastrointestinal symptoms associated with CoVID-19 147.

Pancreatitis and associated fibrosis induced by L-Arginine can be attenuated by sodium butyrate, which reduces collagen deposition and nitric oxide along with inhibition of profibrotic pancreatic stellate cells 148. Butyric acid ameliorates bleomycin induced pulmonary fibrosis by attenuating leukocytes infiltration, oxidative stress and NF-κB activation 149.

Consequently, based on the evidence presented, the potential anti-inflammatory and tissue protective effects of butyric acid on lungs and gut, along with its ability to modulate gut microbiome diversity, enhancing production of endogenous butyric acid could be a better preventive approach to manage CoVID-19 over dexamethasone ( Figure 2). However, there is a need for more detailed studies and clinical trials to determine the potency and long-term effect of butyric acid in the preventive management of seriously ill CoVID-19 patients.
 
Sodium butyrate alleviates cholesterol gallstones by regulating bile acid metabolism


Cholesterol overloading and bile acid metabolic disorders play an important role in the onset of cholesterol gallstone (CGS). Short-chain fatty acids (SCFAs) can regulate bile acid metabolism by modulating the gut microbiota. However, the role and mechanism by which sodium butyrate (NaB) targets bile acids to attenuate CGS are still unknown. In this study, continuous administration of 12 mg/day for 8 weeks was decreased the incidence of gallstones induced by lithogenic diet (LD) from 100% to 25%. NaB modulated SCFAs and improved the gut microbiota. The remodeling of the gut microbiota changed the bile acid compositions and decreased cecal tauro-α-muricholic acid (T-α-MCA) and tauro-β-muricholic acid (T-β-MCA) which are effective farnesoid X receptor (FXR) antagonists. The quantitative real-time PCR examination showed that NaB significantly increased levels of ileal Fxr, fibroblast growth factor-15 (Fgf-15) and small heterodimer partner (Shp) mRNA and subsequently inhibited bile acid synthesis. In addition, NaB enhanced bile acid excretion by increasing the levels of hepatic multidrug resistance protein 2 (Mdr2) and bile salt export pump (Bsep) mRNA, and it enhanced bile acid reabsorption in the intestine by increasing the levels of ileal bile acid transporter (Ibat) mRNA. In addition, NaB reduced the absorption of cholesterol in the intestine and inhibited the excretion of cholesterol in the liver, which reduced the cholesterol concentration in serum and bile. Furthermore, the protective effects of NaB administration were abolished by FXR antagonists.
 

61sO6A3ijKL._AC_AA152_.jpg
21.99 euros​

Hepatica Butyrate de sodium 500mg 90 gélules végétales, 385mg d’acide butyrique​

Faut-il en prendre tout le temps ou en cure de 3 mois ? Merci pour vos précieux conseils.
Should I take it all the time or as a 3-month cure? Thanks for your precious advice.
 
Legumes are one of the few foods with reasonable amounts of protein and fiber, the secret weapons of satiety (study). In addition, its carbohydrate is slow-release, avoiding insulin peaks.

For your intestine, they are a double-edged sword.

On the one hand, they provide fiber and a certain amount of resistant starch. This is beneficial for your microbiota (butyrate production), and also improves insulin sensitivity.

On the other hand, legumes have a high amount of FODMAPs, which is problematic for many people (detail). If you are sensitive to this type of carbohydrate you will notice gas and bloating. If your system doesn't tolerate them well, don't eat them.


So a traditional legume stew to create butyrate a week and a good cured sheep cheese to maintain it.

Practically it was and in many cases is the diet of the people of the small towns of northern Spain. People who easily reach close to 100 years of life.

Very long-lived.
 
Already abounding on the subject, a few months ago I saw in the Covid thread a statistical table of the incidence of mortality and disease derived from vaccines in Europe.

Curiously, Spain and Portugal very close, had a relatively low incidence.

At that time I wondered why this was.

I found it very strange.

The culture of "pottage" of legumes is deeply rooted in the diet and consumption of fermented dairy products (cheese and butter), so perhaps there is a relationship in this.
 
Should I take it all the time or as a 3-month cure?
Start with 3 months and then take a break. You can re-start for the Winter, which is the one time all of us need extra help.
good cured sheep cheese
Even though it might work out for some populations, I would not extrapolate the health of the Mediterranean peoples to the rest of the world. In my experience, sick Spanish people look better than say someone descended from the British Isles or France with the same disease. I would stay from all dairy products. Specially people prone to autoimmune conditions and issues in general, as it is in general the case for a lot of forum members.
 
Even though it might work out for some populations, I would not extrapolate the health of the Mediterranean peoples to the rest of the world. In my experience, sick Spanish people look better than say someone descended from the British Isles or France with the same disease. I would stay from all dairy products. Specially people prone to autoimmune conditions and issues in general, as it is in general the case for a lot of forum members.
Of course.

The point is that for a long time I have been thinking about the good health and longevity of life (in optimal conditions) of these people, whom I know very well by close family in that way of life.

Tough people with very little physical and mental deterioration.

But yes, always watching the autoimmune conditions of each one.
 
Some of the butyrate-producing-bacteria are:

The majority of Firmicutes are butyrate-producing bacteria. At the genus level, Ruminococcus, Clostridium, Eubacterium, and Coprococcus are common butyrate-producing bacteria. Clostridium butyricum (C. butyricum) is relatively common in the Clostridium genus[4]. Others include Faecalibacterium, Butyrivibrio, etc.,[5]. In the genus Eubacterium, Eubacterium Hallii (E. Hallii) and Eubacterium Rectale are among the most abundant butyrate-producing bacterial strains in human feces[6]. Actinomycetes, Bacteroidetes, Proteobacteria, Spirochetes also have been identified as potential butyrate-producing bacteria[2].

Prospects for clinical applications of butyrate-producing bacteria
 
Merci Gaby, quelle chance nous avons de vous avoir toujours disponible pour nous conseiller, MERCI d'être là pour nous...
Soyez bénie... 🥰

Thank you Gaby, how lucky we are to have you always available to advise us, THANK YOU for being there for us...
Be blessed... 🥰
 
Back
Top Bottom