Free fatty acid receptor 3

Last updated
FFAR3
Identifiers
Aliases FFAR3 , FFA3R, GPR41, GPR42, free fatty acid receptor 3
External IDs OMIM: 603821 MGI: 2685324 HomoloGene: 82482 GeneCards: FFAR3
Orthologs
SpeciesHumanMouse
Entrez
Ensembl
UniProt
RefSeq (mRNA)

NM_005304

NM_001033316

RefSeq (protein)

NP_005295

NP_001028488

Location (UCSC) Chr 19: 35.36 – 35.36 Mb Chr 7: 30.55 – 30.56 Mb
PubMed search [3] [4]
Wikidata
View/Edit Human View/Edit Mouse

Free fatty acid receptor 3 (FFAR3, also termed GPR41) protein is a G protein coupled receptor (i.e., GPR or GPCR) that in humans is encoded by the FFAR3 gene (i.e., GPR41 gene). [5] GPRs reside on cell surfaces, bind specific signaling molecules, and thereby are activated to trigger certain functional responses in their parent cells. FFAR3 is a member of the free fatty acid receptor group of GPRs that includes FFAR1 (i.e., GPR40), FFAR2 (i.e., GPR43), and FFAR4 (i.e., GPR120). [6] All of these FFARs are activated by fatty acids. FFAR3 and FFAR2 are activated by certain short-chain fatty acids (SC-FAs), i.e., fatty acids consisting of 2 to 6 carbon atoms [7] whereas FFFAR1 and FFAR4 are activated by certain fatty acids that are 6 to more than 21 carbon atoms long. [8] [9] [10] Hydroxycarboxylic acid receptor 2 is also activated by a SC-FA that activate FFAR3, i.e., butyric acid. [11]

Contents

The human FFAR3 gene is located next to the FFAR2 gene at locus 13.12 on the long (i.e., "q") arm of chromosome 19 (location abbreviated as 19q13.12). The human FFAR3 and FFAR2 proteins consist of 346 and 330 amino acids, respectively, [12] and share about a 40% amino acid sequence homology. [13] The two FFARs have been found to form a heteromer complex (i.e., FFAR3 and FFAR2 bind to each other and are activated together by a SC-FA) in human monocytes, macrophages, and the immortalized embryonic kidney cells, HEK 293 cells. When stimulated by a SC-FA, the cells expressing both FFAR3 and FFAR2 may form this heterodimer and thereby activate cell signaling pathways and mount responses that differ from those of cells expressing only one of these FFARs. [14] The formation of GPR43-GPR41 heterodimers has not been evaluated in most studies and may explain otherwise conflicting results on the roles of FFAR3 and FFAR2 in cell function. [10] [15] [16] Furthermore, SC-FAs can alter the function of cells independently of FFAR3 and FFAR2 by altering the activity of cellular histone deacetylases which regulate the transcription of various genes or by altering metabolic pathways which alter cell functions. [17] [18] Given these alternate ways for SC-FAs to activate cells as wells at the ability of SC-FAs to activate FFAR2 or, in the case of butyric acid, hydroxycarboxylic acid receptor 2, the studies reported here focus on those showing that the examined action(s) of an SC-FA is absent or reduced in cells, tissues, or animals that have no or reduced FFAR3 activity due respectively to knockout (i.e., removal or inactivation) or knockdown (i.e., reduction) of the FFAR3 protein gene, i.e., the Ffar3 gene in animals or FFAR3 gene in humans.

Certain bacteria in the gastrointestinal tract ferment fecal fiber into SC-FAs and excrete them as waste products. The excreted SC-FAs enter the gastrointestinal walls, diffuse into the portal venous system, and ultimately flow into the systemic circulation. During this passage, they can activate the FFAR3 on cells in the intestinal wall as well as throughout the body. [19] This activation may: suppress the appetite for food and thereby reduce overeating and the development of obesity; [20] [21] inhibit the liver's accumulation of fatty acids and thereby the development of fatty liver diseases; [22] decrease blood pressure and thereby the development of hypertension and hypertension-related cardiac diseases; [23] modulate insulin secretion and thereby the development and/or symptoms of type 2 diabetes; [24] reduce heart rate and blood plasma norepinephrine levels and thereby lower total body energy expenditures; [19] and suppress or delay the development of allergic asthma. [25]

The specific types of bacteria in the intestines can be modified to increase the number which make SC-FAs by using foods that stimulate the growth of these bacteria (i.e., prebiotics), preparations of SC-FA-producing bacteria (i.e., probiotics), or both methods (see synbiotics). [26] Individuals with disorders that are associated with low levels of the SC-FA-producing intestinal bacteria may show improvements in their conditions when treated with prebiotics, probiotics, or synbiotics while individuals with disorders associated with high levels of SC-FAs may show improvements in their conditions when treated with methods, e.g., antibiotics, that reduce the intestinal levels of these bacteria. [19] [27] (For information on these treatments see Disorders treated by probiotics and Disorders treated by prebiotics). In addition, drugs are being tested for their ability to act more usefully, potently, and effectively than SC-FAs in stimulating or inhibiting FFAR3 and thereby for treating the disoders that are inhibited or stimulated, respectively, by SC-FAs. [28]

FFAR3-activating and inhibiting agents

The SC-FAs that activate FFAR3 include proprionic, butyric, acetic, [29] valeric [19] caproic, [27] and formic acids. [30] (Confusingly, butyric acid also activates hydroxycarboxylic acid receptor 2 [30] and β-hydroxybutyric acid has been reported to stimulate or inhibit FFAR3. [31] ) FFAR2 is activated by many of these same SC-FAs but differs from FFAR3 in its relative binding affinities for them. In humans, the binding affinity ranking of FFAR3 is: propionic = butyric = valeric > acetic > formic acids (acetic [29] and formic [30] acids have very low binding affinities for, and therefore must be at extremely high levels to activate, FFAR3); FFAR2's relative binding affinity ranking for these SC-FAs is: acetic = propionic > butyric > valeric = formic acids. [30] AR420626 (also termed 1-MCPC [12] or its chemical name, [(S)-2-(4-chlorophenyl)-3,3-dimethyl-N-(5-phenylthiazol-2-yl)butamide (CFMB)/AR420626] [32] ) has been reported to be a selective activator of FFAR3 [33] but has also been reported to activate FFAR2 [32] and to inhibit the activation of FFAR3. [19] Its actions require further characterizations. [19] AR399519 and CF3-MQC have been reported to inhibit the activation of mouse FFAR3; the actions of these agents also require further characterizations. [12]

Tissues and cells expressing FFAR3

Studies have reported that humans express FFAR3 in their: a) enteroendocrine L cells and K cells of the intestines; [10] b) endothelium of blood vessels in the frontal cortex of the brain, [34] pancreatic β-cells, [35] and adipose. i.e., fat, tissue (but not in mouse adipose tissue); [36] c) the vascular endothelium of the myometrium, the epithelium of the amnion, chorion and placenta, and certain immune cells in these tissues of pregnant women; [37] d) the hippocampus of the brain; [38] e) sympathetic ganglia, i.e., autonomic ganglia of the sympathetic nervous system; [12] [39] f) certain types of immune cells, i.e., blood monocytes (but not mouse monocytes), basophils, [18] [40] [41] dendritic cells derived from human monocytes isolated from whole blood, and the tissues containing these blood cells, i.e., the bone marrow, spleen, lymph nodes, and thymus; [42] and g) alveolar macrophages, and macrophages in various other tissues; [18] and h) certain immortalised cell lines, i.e., MCF-7 breast cancer, [43] HCT116 colorectal cancer, [44] HEK293 embryonic kidney, [14] [41] U937 leukemic promonocyte, THP-1 leukemic monocyte, EoL-1 leukemic eosinophil, Jurcat leukemic T lymphocyte, MOLT-4 T lymphoblast leukemic, and HL60 acute myeloid leukemia cells (but only when the HL60 cells are pre-treated with phorbol 12-myristate 13-acetate to promote their cellular differentiation). [41] As noted, the expression of FFAR3 in the cells and tissues of animals are not always the same as those in humans.

Functions

Regulation of satiety and obesity

L cells are enteroendocrine cells, i.e., specialized cells that secrete hormones directly into the circulation. L cells reside in the epithelium of the gastrointestinal tract, particularly the terminal ileum and colon. They are stimulated to secrete PYY (also termed peptide YY) and GLP-1 (also termed glucagon-like peptide-1) by the SC-FAs that accumulate inside the intestines after feeding. L cells express FFAR3 and/or FFAR2. [29] Ffar3 and Ffar2 gene knock out mice show reduced secretions of GLP-1 and PYY. [45] Studies have shown that Ffar3 gene knockout mice fed a high fat diet have significant increases in their food intake and body weights compared to wild-type (i.e., genetically unaltered) mice. [19] These and other studies in animals suggest that the activation of FFAR3 and FFAR2 on L cells by SC-FAs triggers the release of PYY and GLP-1 both of which, among various other activities, inhibit gastric emptying and thereby suppress appetite and the development of obesity. [19] [21] Further studies are needed to determine if FFAR3 plays a similar role in human satiety and obesity. [19] It should be noted, however, that Semaglutide, also called Wegovy, is a peptide with a modified GLP-1-like structure. It strongly stimulates GLP-1 receptors and thereby suppresses appetite and promotes weight loss in obese individuals. [46]

Leptin is a peptide hormone released by adipose tissue that triggers satiety and thereby tends to reduce or stop further food intake and the development of obesity. It also plays a role in female reproductive function, lipolysis (e.g., the breakdown of triglycerides into their component free fatty acids and glycerol), the growth of fetuses, inflammation, and angiogenesis (i.e., the formation of new blood vessels from pre-existing blood vessels). [20] While studies have suggested that the SC-FA-induced activation of FFAR3 leads to the secretion of leptin from the white adipose tissue of intact animals and the fat tissue isolated from human tissues, other studies have suggested that FFAR2 rather than FFAR3 is responsible for the SC-FA-induced release of leptin form fat tissue. A systematic review of the published studies on this issue concluded that SC-FA-induced activation of FFAR3 is likely responsible for the SC-FA-induced release of leptin from cultured fat tissue taken from animals. However, the data were insufficient to support a role for FFAR3 in the release of leptin from cultured human fat tissues or the fat tissue of intact animals. The role of FFAR3 stimulation of leptin release in apatite suppression and obesity needs further study. [47]

Regulation of liver fatty acid storage and fatty liver disease

In a high-fat diet-induced obesity model of fatty liver disease (i.e., excessive buildup of fat in the liver), mice fed a diet that increased intestinal levels of SC-FAs showed reductions in their livers' synthesis of lipids, triglyceride levels, and weights. These reductions did not occur in Ffar3 gene knockout mice but did occur in Ffar2 gene knocked-out mice. These results indicate that the SC-FA-induced activation of FFAR3 suppresses the liver's accumulation of fatty acids that underlies the development of fatty liver disease in this mouse model. [22] Other studies have found that Ffar3 gene knockout mice showed less weight gain than wild-type mice under standard laboratory conditions but this difference was lost in mice reared under germ-free conditions (i.e., which causes the mice to have lower intestinal and tissue levels of SC-FAs). [13] These findings indicated that the activation of FFAR3 but not FFAR2 by SC-FAs protects against developing fatty liver disease in mice; [13] [30] they also support studies to determine if FFAR3 has these actions in humans and if FFAR3 activators can be used to treat or prevent human fatty liver diseases such as non-alcoholic fatty liver disease. [48]

Regulation of insulin secretion and diabetes

Individuals with type 2 diabetes, which accounts for 90% of all diabetes cases, have decreases in the proliferation, maturation, and activity of their pancreatic islet insulin-secreting beta-cells as well as the potency of insulin's actions. These decreases result in reduced insulin secretion, hyperglycemia, and the many other afflictions associated with this disorder. Studies in the past have reported that the activation of FFAR3 reduced the insulin secreted by 1) human and mouse beta cells in vivo, 2) cultured human and murine beta cell-containing pancreatic islets, and 3) cultured beta cell lines. [12] [24] These studies showed that acetic acid-induced inhibition of insulin secretion by mouse pancreatic islets did not occur in islets that had both of their Ffar3 and Ffar2 genes knocked out but had no effect on insulin secretion in islets that had only one of the two genes knocked out. [24] In contrast to this, however, a recent study of streptozotocin-induced and high-fat diet-induced murine models of diabetes found that the FFAR3-activating drug, AR420626, increased blood plasma insulin levels and stimulated skeletal muscle to take up glucose and thereby improved glucose tolerance test results. [49] Other recent studies have reported that activated FFAR3 may reduce, increase, or have little effect on insulin secretion depending on 1) the levels of ambient glucose and FFAR3 activators studied, 2) human or animal species studied, 3) age of the animals studied, and 4) variations in the proportions of alpha, beta, and delta cells in the pancreatic islets of humans. The role of FFAR3 in human as well as animal models of insulin secretion and diabetes requires further studies. [35]

Regulation of blood pressure and hypertension-induced cardiovascular disorders

An early study showed that the intravenous injection of propionic acid into mice induced a brief (<5 min) hypotensive response as defined by drops in their mean arterial pressures. This response was reduced in mice that had one of their two Ffar3 genes knocked out and absent in mice that had both Ffar3 genes knocked out. [23] [50] A subsequent study reported that Ffar3 gene knockout mice developed abnormally high diastolic blood pressuress and pulse pressures (i.e., systolic minus diastolic blood pressures) as well as increased amounts of cardiac collagen and elastin connective tissue and increased cardiac stiffness as evidenced by a reduce rate of heart muscle relaxation measured by pressure-volume loop analysis tau levels. [51] [52] Taken together, these studies suggest that in mice SC-FA-activated FFAR3 functions to reduce blood pressure as well as some of the consequences of hypertension such as cardiac fibroses and poor myocardial contractility. [53] Studies in humans have found that individuals undergoing hemodialysis using dialysis solutions that contain acetic acid often develop hypotension; the role of FFAR3 in this response, if any, was not investigated. [23] [50] And, a study of 69 individuals (55.1% women, mean age 59.8 years) found that arterial stiffness was associated with lower levels of FFAR3 and FFAR2 in circulating blood immune cells (particularly regulatory T cells which are known to be protective in murine models of hypertension [54] ). [52] Overall. the mouse studies suggest that FFAR3 contributes to suppressing hypertension and its subsequent effects on the heart in mice [51] and that that SC-FA-activated FFAR3 and/or FFAR2 may have vasodilatory actions and thereby suppress the development of hypertension and hypertension-induced arterial stiffness in humans. Further studies in humans are needed to investigate the latter possibilities. [51] [54]

Regulation of the sympathetic nervous system and its control of heart rate and energy expenditure

Studies have shown that compared to wild type mice, Ffar3 gene knockout mice have: a) significantly smaller-sized sympathetic nervous system ganglia as judged by measurements of this systems' largest ganglia, the superior cervical ganglion; b) significantly slower heart rates; and c) significantly lower norepinephrine levels in their blood plasma. (Norepinephrine is a neurotransmitter that is released by sympathetic nervous system neurons and among other actions increases heart rate and total body energy expenditure.) Furthermore, the treatment of wild type mice with propionic acid increased their heart rates but did not do so in Ffar3 gene knockout mice. [19] [39] Finally, the offspring of Ffar3-gene knockout mice had slower heart rates (as well as lower body temperatures) than the offspring of wild type mice. [10] These findings indicate that FFAR3 regulates heart rates and energy expenditure in mice. Studies are needed to determine if it does so in humans. [19] [39]

Regulation of allergic asthma reactions

Asthma may be atopic (i.e., symptoms triggered by allergens) or non-atopic (i.e., symptoms triggered by non-allergenic factors such as cold air). The studies reported here relate to allergen-induced asthma. Mice fed a diet that lowers their SC-FAs levels and then given intranasal injections of dust mite extract developed dust mite allergy asthma reactions to the injections. Their respiratory tract airways had increased numbers of eosinophils and goblet cells as well as excessive levels of mucus; their lung tissue levels of interleukin-4, interleukin-5, interleukin-13, and interleukin-17A and serum immunoglobulin E levels were elevated; and their airway resistance response to a bronchial challenge test was high. In contrast, mice fed a diet that increased their SC-FAs levels developed less of these responses to the extract. Furthermore, mice on the SC-FA lowering diet that were given propionic acid also had far less of these responses to the mite extract. And, Ffar3 (but not Ffar2) gene knockout mice on the low SF-FA diet did not show rises in their lung airway eosinophil levels in response to the mite extract (this was the only parameter of asthma reported in the knockout studies). These finding implicate propionic acid and FFAR3 in the suppression of asthma allergic reactions to mite extract in mice. [55]

A second study investigated the effects that an inulin-rich diet (which raises bodily SC-FA levels) feed to rats had on their offsprings' development of asthma. Pregnant rats were feed a normal or inulin-rich diet for 1 week. Their offspring were injected with ovalbumin on days 21 and 29 after birth, 7 days later challenged with aerosolized ovalbumin, and on the next day examined for their responses to the aerosol. Compared to the offspring of mothers on a normal diet, the offspring of mothers on the inulin diet had lower levels of lung inflammatory cells, less histological evidence of allergic lung disease, lower lung tissue levels of immunoglobulin E, interleukin-4, and interleukin-17, and significantly elevated lung levels of FFAR3 (Lung FFAR2 levels were not significantly elevated). These results indicate that a diet promoting the production of SC-FAs in pregnant rats suppresses the development of asthmatic disease in their offspring; this suppression may involve FFAR3. [25] In a similar study, newborn mice were feed breast milk from mothers who had drunk pure water or water containing a SC-FA. After 3 weeks, the newborns were weaned off the mothers' milk, feed plain water, and 3 weeks thereafter sensitized to and challenged by injection of mite extract into their tracheas. Mothers who drank pure water or water laced with acetic or butyric acid and sensitized to the mite extract had asthma signs after challenge with the extract whereas mothers who drank propionic acid-laced water had far less of these signs. Furthermore, Ffar3 gene knockout mothers who drank propionic acid-laced water and then sensitized to the mite extract had asthma signs similar to these in wild type mothers challenged with the extract. The study also found that the offspring of mothers who drank propionic acid-laced water had fewer eosinophils and T helper cells in their airways than the offsprings of mothers who drank pure water, acetic acid-laced water, or butyric acid-laced water. Propionic acid-laced water did not suppress the development of asthma in Ffar3 gene knockout offsprings. These results indicate that ingestion of propionic acid, but not acetic or butyric acid, suppresses the development of allergic asthma in adult as well as newborn rats and does so by a FFAR3-dependent mechanism. The studies also indicate that the milk of pregnant rats who consumed propionic acid-laced but not those who drank pure water reduced the susceptibility of newborn rats to developing allergic asthma by a mechanism dependent on FFAR3 in the mothers as well as the offsprings. These findings support further studies to determine if propionic acid or other FFAR3 activators would be useful for preventing and/or treating asthma in humans. [18] [56]

A study of humans living on European farms or in non-farm rural areas reported that the fecal levels of butyric but not acetic acid in 12 month old children who had not develop asthma by the time they entered the first year of school were significantly higher than these levels in children who did develop asthma by the school entry age. [57] A study conducted in Canada reported that the levels of fecal acetic acid (but not butyric or propionic acid) were lower in 3 month old human infants who were predicted to have asthma by school age (based on a Phylogenetic Investigation of Communities by Reconstruction of Unobserved States prediction algorithm) compared to infants predicted not to do so. [58] Finally, a study conducted in Japan found that the fecal levels of propionic but not acetic or butyric acid trended lower in 1 month old human infants that developed asthma by age 5 than in infants that did not develop asthma. The fecal levels of propionic as well acetic and butyric acid obtained from 1 week-, 1 year, and 5-year-old infants did not show this trend. [56] The different SC-FA implicated in suppressing asthma in these three studies may reflect dietary or other differences between the populations of the three countries. In all events, the studies allow that, based on rodent studies, FFAR3 may mediate these SG-FA actions and, based on human studies, SC-FAs may act to suppress, or at least delay) the onset of, asthma in children Further studies are needed to determine if FFAR3 is involved in the apparent actions of the cited SC-FAs in the development of asthma in children. [56]

See also

Related Research Articles

<span class="mw-page-title-main">Butyric acid</span> Chemical compound

Butyric acid, also known under the systematic name butanoic acid, is a straight-chain alkyl carboxylic acid with the chemical formula CH3CH2CH2CO2H. It is an oily, colorless liquid with an unpleasant odor. Isobutyric acid is an isomer. Salts and esters of butyric acid are known as butyrates or butanoates. The acid does not occur widely in nature, but its esters are widespread. It is a common industrial chemical and an important component in the mammalian gut.

<span class="mw-page-title-main">Indole-3-acetic acid</span> Chemical compound

Indole-3-acetic acid is the most common naturally occurring plant hormone of the auxin class. It is the best known of the auxins, and has been the subject of extensive studies by plant physiologists. IAA is a derivative of indole, containing a carboxymethyl substituent. It is a colorless solid that is soluble in polar organic solvents.

<span class="mw-page-title-main">Thromboxane receptor</span> Mammalian protein found in Homo sapiens

The thromboxane receptor (TP) also known as the prostanoid TP receptor is a protein that in humans is encoded by the TBXA2R gene, The thromboxane receptor is one among the five classes of prostanoid receptors and was the first eicosanoid receptor cloned. The TP receptor derives its name from its preferred endogenous ligand thromboxane A2.

Arachidonate 5-lipoxygenase, also known as ALOX5, 5-lipoxygenase, 5-LOX, or 5-LO, is a non-heme iron-containing enzyme that in humans is encoded by the ALOX5 gene. Arachidonate 5-lipoxygenase is a member of the lipoxygenase family of enzymes. It transforms essential fatty acids (EFA) substrates into leukotrienes as well as a wide range of other biologically active products. ALOX5 is a current target for pharmaceutical intervention in a number of diseases.

Free fatty acid receptors (FFARs) are G-protein coupled receptors (GPRs). GPRs are a large family of receptors. They reside on their parent cells' surface membranes, bind any one of a specific set of ligands that they recognize, and thereby are activated to elicit certain types of responses in their parent cells. Humans express more than 800 different types of GPCRs. FFARs are GPCR that bind and thereby become activated by particular fatty acids. In general, these binding/activating fatty acids are straight-chain fatty acids consisting of a carboxylic acid residue, i.e., -COOH, attached to aliphatic chains, i.e. carbon atom chains of varying lengths with each carbon being bound to 1, 2 or 3 hydrogens. For example, propionic acid is a short-chain fatty acid consisting of 3 carbons (C's), CH3-CH2-COOH, and docosahexaenoic acid is a very long-chain polyunsaturated fatty acid consisting of 22 C's and six double bonds : CH3-CH2-CH1=CH1-CH2-CH1=CH1-CH2-CH1=CH1-CH2-CH1=CH1-CH2-CH1=CH1-CH2-CH1=CH1-CH2-CH2-COOH.

<span class="mw-page-title-main">ALOX15</span> Lipoxygenase found in humans

ALOX15 is, like other lipoxygenases, a seminal enzyme in the metabolism of polyunsaturated fatty acids to a wide range of physiologically and pathologically important products. ▼ Gene Function

<span class="mw-page-title-main">Free fatty acid receptor 1</span> Protein-coding gene in the species Homo sapiens

Free fatty acid receptor 1 (FFAR1), also known as G-protein coupled receptor 40 (GPR40), is a rhodopsin-like G-protein coupled receptor that is coded by the FFAR1 gene. This gene is located on the short arm of chromosome 19 at position 13.12. G protein-coupled receptors reside on their parent cells' surface membranes, bind any one of the specific set of ligands that they recognize, and thereby are activated to trigger certain responses in their parent cells. FFAR1 is a member of a small family of structurally and functionally related GPRs termed free fatty acid receptors (FFARs). This family includes at least three other FFARs viz., FFAR2, FFAR3, and FFAR4. FFARs bind and thereby are activated by certain fatty acids.

<span class="mw-page-title-main">GPR42</span> Protein-coding gene in the species Homo sapiens

Putative G-protein coupled receptor 42 is a protein that in humans is encoded by the GPR42 gene. The human GPR gene is located at the same site as the human FFAR1, FFAR, and FFAR3 genes, i.e., on the long arm of chromosome 19 at position 23.33. This gene appears to be a segmental duplication of the FFAR3 gene. The human GPR42 gene codes for several proteins with a FFAR3-like structure but their expression in various cell types and tissues as well as their activities and functions have not yet been clearly defined in any scientific publication followed by PubMed as of 2023.

<span class="mw-page-title-main">Free fatty acid receptor 2</span> Protein-coding gene in the species Homo sapiens

Free fatty acid receptor 2 (FFAR2), also termed G-protein coupled receptor 43 (GPR43), is a rhodopsin-like G-protein coupled receptor. It is coded by the FFAR2 gene. In humans, the FFAR2 gene is located on the long arm of chromosome 19 at position 13.12. Like other GPCRs, FFAR2s reside on the surface membrane of cells and when bond to one of their activating ligands regulate the function of their parent cells. FFAR2 is a member of a small family of structurally and functionally related GPRs termed free fatty acid receptors (FFARs). This family includes three other receptors which, like FFAR2, are activated by certain fatty acids: FFAR1, FFAR3 (GPR41), and FFAR4 (GPR120). FFAR2 and FFAR3 are activated by short-chain fatty acids whereas FFAR1 and FFAR4 are activated by long-chain fatty acids.

<span class="mw-page-title-main">OXGR1</span> Protein-coding gene in the species Homo sapiens

2-Oxoglutarate receptor 1 (OXGR1), also known as cysteinyl leukotriene receptor E (CysLTE) and GPR99, is a protein that in humans is encoded by the OXGR1 gene. The Gene has recently been nominated as a receptor not only for 2-oxoglutarate but also for the three cysteinyl leukotrienes (CysLTs), particularly leukotriene E4 (LTE4) and to far lesser extents LTC4 and LTD4. Recent studies implicate GPR99 as a cellular receptor which is activated by LTE4 thereby causing these cells to contribute to mediating various allergic and hypersensitivity responses.

<span class="mw-page-title-main">GPR132</span> Protein-coding gene in the species Homo sapiens

G protein coupled receptor 132, also termed G2A, is classified as a member of the proton sensing G protein coupled receptor (GPR) subfamily. Like other members of this subfamily, i.e. GPR4, GPR68 (OGR1), and GPR65 (TDAG8), G2A is a G protein coupled receptor that resides in the cell surface membrane, senses changes in extracellular pH, and can alter cellular function as a consequence of these changes. Subsequently, G2A was suggested to be a receptor for lysophosphatidylcholine (LPC). However, the roles of G2A as a pH-sensor or LPC receptor are disputed. Rather, current studies suggest that it is a receptor for certain metabolites of the polyunsaturated fatty acid, linoleic acid.

<span class="mw-page-title-main">Hydroxycarboxylic acid receptor 2</span> Protein-coding gene in the species Homo sapiens

Hydroxycarboxylic acid receptor 2 (HCA2), also known as GPR109A and niacin receptor 1 (NIACR1), is a protein which in humans is encoded (its formation is directed) by the HCAR2 gene and in rodents by the Hcar2 gene. The human HCAR2 gene is located on the long (i.e., "q") arm of chromosome 12 at position 24.31 (notated as 12q24.31). Like the two other hydroxycarboxylic acid receptors, HCA1 and HCA3, HCA2 is a G protein-coupled receptor (GPCR) located on the surface membrane of cells. HCA2 binds and thereby is activated by D-β-hydroxybutyric acid (hereafter termed β-hydroxybutyric acid), butyric acid, and niacin (also known as nicotinic acid). β-Hydroxybutyric and butyric acids are regarded as the endogenous agents that activate HCA2. Under normal conditions, niacin's blood levels are too low to do so: it is given as a drug in high doses in order to reach levels that activate HCA2.

<span class="mw-page-title-main">Free fatty acid receptor 4</span> Protein-coding gene in the species Homo sapiens

Free Fatty acid receptor 4 (FFAR4), also termed G-protein coupled receptor 120 (GPR120), is a protein that in humans is encoded by the FFAR4 gene. This gene is located on the long arm of chromosome 10 at position 23.33. G protein-coupled receptors reside on their parent cells' surface membranes, bind any one of the specific set of ligands that they recognize, and thereby are activated to trigger certain responses in their parent cells. FFAR4 is a rhodopsin-like GPR in the broad family of GPRs which in humans are encoded by more than 800 different genes. It is also a member of a small family of structurally and functionally related GPRs that include at least three other free fatty acid receptors (FFARs) viz., FFAR1, FFAR2, and FFAR3. These four FFARs bind and thereby are activated by certain fatty acids.

<span class="mw-page-title-main">Oxoeicosanoid receptor 1</span> Protein-coding gene in the species Homo sapiens

Oxoeicosanoid receptor 1 (OXER1) also known as G-protein coupled receptor 170 (GPR170) is a protein that in humans is encoded by the OXER1 gene located on human chromosome 2p21; it is the principal receptor for the 5-Hydroxyicosatetraenoic acid family of carboxy fatty acid metabolites derived from arachidonic acid. The receptor has also been termed hGPCR48, HGPCR48, and R527 but OXER1 is now its preferred designation. OXER1 is a G protein-coupled receptor (GPCR) that is structurally related to the hydroxy-carboxylic acid (HCA) family of G protein-coupled receptors whose three members are HCA1 (GPR81), HCA2, and HCA3 ; OXER1 has 30.3%, 30.7%, and 30.7% amino acid sequence identity with these GPCRs, respectively. It is also related to the recently defined receptor, GPR31, for the hydroxyl-carboxy fatty acid 12-HETE.

Prostaglandin EP<sub>3</sub> receptor Protein-coding gene in the species Homo sapiens

Prostaglandin EP3 receptor (53kDa), also known as EP3, is a prostaglandin receptor for prostaglandin E2 (PGE2) encoded by the human gene PTGER3; it is one of four identified EP receptors, the others being EP1, EP2, and EP4, all of which bind with and mediate cellular responses to PGE2 and also, but generally with lesser affinity and responsiveness, certain other prostanoids (see Prostaglandin receptors). EP has been implicated in various physiological and pathological responses.

<span class="mw-page-title-main">Prostacyclin receptor</span> Mammalian protein found in Homo sapiens

The Prostacyclin receptor, also termed the prostaglandin I2 receptor or just IP, is a receptor belonging to the prostaglandin (PG) group of receptors. IP binds to and mediates the biological actions of prostacyclin (also termed Prostaglandin I2, PGI2, or when used as a drug, epoprostenol). IP is encoded in humans by the PTGIR gene. While possessing many functions as defined in animal model studies, the major clinical relevancy of IP is as a powerful vasodilator: stimulators of IP are used to treat severe and even life-threatening diseases involving pathological vasoconstriction.

<span class="mw-page-title-main">Sodium-coupled monocarboxylate transporter 1</span> Protein-coding gene in the species Homo sapiens

Sodium-coupled monocarboxylate transporter 1 (i.e., SMCT1) and sodium-coupled monocarboxylate transporter 2 (i.e., SMCT2) are plasma membrane transport proteins in the solute carrier family. They transport sodium cations in association with the anionic forms (see conjugated base) of certain short-chain fatty acids (i.e., SC-FAs) through the plasma membrane from the outside to the inside of cells. For example, propionic acid (i.e., CH
3
CH
2
CO
2
H
) in its anionic "propionate" form (i.e., CH
3
CH
2
CO
2
) along with sodium cations (i.e., Na+) are co-transported from the extracellular fluid into a SMCT1-epxressing cell's cytoplasm. Monocarboxylate transporters (MCTs) are also transport proteins in the solute carrier family. They co-transport the anionic forms of various compounds into cells in association with proton cations (i.e. H+). Four of the 14 MCTs, i.e. SLC16A1 (i.e., MCT1), SLC16A7 (i.e., MCT22), SLC16A8 (i.e., MCT3), and SLC16A3 (i.e., MCT4), transport some of the same SC-FAs anions that the SMCTs transport into cells. SC-FAs do diffuse into cells independently of transport proteins but at the levels normally occurring in tissues far greater amounts of the SC-FAs are brought into cells that express a SC-FA transporter.

<span class="mw-page-title-main">12-Hydroxyheptadecatrienoic acid</span> Chemical compound

12-Hydroxyheptadecatrienoic acid (also termed 12-HHT, 12(S)-hydroxyheptadeca-5Z,8E,10E-trienoic acid, or 12(S)-HHTrE) is a 17 carbon metabolite of the 20 carbon polyunsaturated fatty acid, arachidonic acid. It was discovered and structurally defined in 1973 by P. Wlodawer, Bengt I. Samuelsson, and M. Hamberg, as a product of arachidonic acid metabolism made by microsomes (i.e. endoplasmic reticulum) isolated from sheep seminal vesicle glands and by intact human platelets. 12-HHT is less ambiguously termed 12-(S)-hydroxy-5Z,8E,10E-heptadecatrienoic acid to indicate the S stereoisomerism of its 12-hydroxyl residue and the Z, E, and E cis-trans isomerism of its three double bonds. The metabolite was for many years thought to be merely a biologically inactive byproduct of prostaglandin synthesis. More recent studies, however, have attached potentially important activity to it.

<span class="mw-page-title-main">13-Hydroxyoctadecadienoic acid</span> Chemical compound

13-Hydroxyoctadecadienoic acid (13-HODE) is the commonly used term for 13(S)-hydroxy-9Z,11E-octadecadienoic acid (13(S)-HODE). The production of 13(S)-HODE is often accompanied by the production of its stereoisomer, 13(R)-hydroxy-9Z,11E-octadecadienoic acid (13(R)-HODE). The adjacent figure gives the structure for the (S) stereoisomer of 13-HODE. Two other naturally occurring 13-HODEs that may accompany the production of 13(S)-HODE are its cis-trans (i.e., 9E,11E) isomers viz., 13(S)-hydroxy-9E,11E-octadecadienoic acid (13(S)-EE-HODE) and 13(R)-hydroxy-9E,11E-octadecadienoic acid (13(R)-EE-HODE). Studies credit 13(S)-HODE with a range of clinically relevant bioactivities; recent studies have assigned activities to 13(R)-HODE that differ from those of 13(S)-HODE; and other studies have proposed that one or more of these HODEs mediate physiological and pathological responses, are markers of various human diseases, and/or contribute to the progression of certain diseases in humans. Since, however, many studies on the identification, quantification, and actions of 13(S)-HODE in cells and tissues have employed methods that did not distinguish between these isomers, 13-HODE is used here when the actual isomer studied is unclear.

Sodium-coupled monocarboxylate transporter 2 (i.e., SMCT2, also termed SLC5A12) is a plasma membrane transport protein in the solute carrier family. It transports sodium cations (i.e., Na+) in association with the anionic forms (see conjugated base) of certain short-chain fatty acids (i.e., SC-FAs) and other agents through the plasma membrane from the outside to the inside of cells. The only other member of the sodium-coupled monocarboxylate transporter group (sometimes referred to as the SLC5A family), SMCT1, similarly co-transports SC-FAs and other agents into cells. Monocarboxylate transporters (MCTs) are also transport proteins in the solute carrier family. They co-transport the anionic forms of various compounds into cells in association with hydrogen cations (i.e. H+). Four of the 14 MCTs, i.e. SLC16A1 (i.e., MCT1), SLC16A7 (i.e., MCT22), SLC16A8 (i.e., MCT3), and SLC16A3 (i.e., MCT4), transport some of the same SC-FAs anions that the two SMCTs transport into cells. SC-FAs do diffuse into cells independently of transport proteins but at the levels normally occurring in tissues greater amounts of the SC-FAs are brought into cells that express a SC-FA transporter.

References

  1. 1 2 3 GRCh38: Ensembl release 89: ENSG00000185897 - Ensembl, May 2017
  2. 1 2 3 GRCm38: Ensembl release 89: ENSMUSG00000019429 - Ensembl, May 2017
  3. "Human PubMed Reference:". National Center for Biotechnology Information, U.S. National Library of Medicine.
  4. "Mouse PubMed Reference:". National Center for Biotechnology Information, U.S. National Library of Medicine.
  5. "Entrez Gene: FFAR3 free fatty acid receptor 3".
  6. Sawzdargo M, George SR, Nguyen T, Xu S, Kolakowski LF, O'Dowd BF (October 1997). "A cluster of four novel human G protein-coupled receptor genes occurring in close proximity to CD22 gene on chromosome 19q13.1". Biochemical and Biophysical Research Communications. 239 (2): 543–7. doi:10.1006/bbrc.1997.7513. PMID   9344866.
  7. Karmokar PF, Moniri NH (December 2022). "Oncogenic signaling of the free-fatty acid receptors FFA1 and FFA4 in human breast carcinoma cells". Biochemical Pharmacology. 206: 115328. doi:10.1016/j.bcp.2022.115328. PMID   36309079. S2CID   253174629.
  8. Briscoe CP, Tadayyon M, Andrews JL, Benson WG, Chambers JK, Eilert MM, Ellis C, Elshourbagy NA, Goetz AS, Minnick DT, Murdock PR, Sauls HR, Shabon U, Spinage LD, Strum JC, Szekeres PG, Tan KB, Way JM, Ignar DM, Wilson S, Muir AI (March 2003). "The orphan G protein-coupled receptor GPR40 is activated by medium and long chain fatty acids". The Journal of Biological Chemistry. 278 (13): 11303–11. doi: 10.1074/jbc.M211495200 . PMID   12496284.
  9. Tunaru S, Bonnavion R, Brandenburger I, Preussner J, Thomas D, Scholich K, Offermanns S (January 2018). "20-HETE promotes glucose-stimulated insulin secretion in an autocrine manner through FFAR1". Nature Communications. 9 (1): 177. Bibcode:2018NatCo...9..177T. doi:10.1038/s41467-017-02539-4. PMC   5766607 . PMID   29330456.
  10. 1 2 3 4 Grundmann M, Bender E, Schamberger J, Eitner F (February 2021). "Pharmacology of Free Falsoatty Acid Receptors and Their Allosteric Modulators". International Journal of Molecular Sciences. 22 (4). doi: 10.3390/ijms22041763 . PMC   7916689 . PMID   33578942.
  11. Offermanns S (March 2017). "Hydroxy-Carboxylic Acid Receptor Actions in Metabolism". Trends in Endocrinology and Metabolism: TEM. 28 (3): 227–236. doi:10.1016/j.tem.2016.11.007. PMID   28087125. S2CID   39660018.
  12. 1 2 3 4 5 Mishra SP, Karunakar P, Taraphder S, Yadav H (June 2020). "Free Fatty Acid Receptors 2 and 3 as Microbial Metabolite Sensors to Shape Host Health: Pharmacophysiological View". Biomedicines. 8 (6): 154. doi: 10.3390/biomedicines8060154 . PMC   7344995 . PMID   32521775.
  13. 1 2 3 Secor JD, Fligor SC, Tsikis ST, Yu LJ, Puder M (2021). "Free Fatty Acid Receptors as Mediators and Therapeutic Targets in Liver Disease". Frontiers in Physiology. 12: 656441. doi: 10.3389/fphys.2021.656441 . PMC   8058363 . PMID   33897464.
  14. 1 2 Ang Z, Xiong D, Wu M, Ding JL (January 2018). "FFAR2-FFAR3 receptor heteromerization modulates short-chain fatty acid sensing". FASEB Journal. 32 (1): 289–303. doi: 10.1096/fj.201700252RR . PMC   5731126 . PMID   28883043.
  15. Martin-Gallausiaux C, Marinelli L, Blottière HM, Larraufie P, Lapaque N (February 2021). "SCFA: mechanisms and functional importance in the gut". The Proceedings of the Nutrition Society. 80 (1): 37–49. doi: 10.1017/S0029665120006916 . PMID   32238208. S2CID   214772999.
  16. Ang Z, Er JZ, Tan NS, Lu J, Liou YC, Grosse J, Ding JL (September 2016). "Human and mouse monocytes display distinct signalling and cytokine profiles upon stimulation with FFAR2/FFAR3 short-chain fatty acid receptor agonists". Scientific Reports. 6: 34145. Bibcode:2016NatSR...634145A. doi:10.1038/srep34145. PMC   5036191 . PMID   27667443.
  17. Carretta MD, Quiroga J, López R, Hidalgo MA, Burgos RA (2021). "Participation of Short-Chain Fatty Acids and Their Receptors in Gut Inflammation and Colon Cancer". Frontiers in Physiology. 12: 662739. doi: 10.3389/fphys.2021.662739 . PMC   8060628 . PMID   33897470.
  18. 1 2 3 4 Tan JK, Macia L, Mackay CR (February 2023). "Dietary fiber and SCFAs in the regulation of mucosal immunity". The Journal of Allergy and Clinical Immunology. 151 (2): 361–370. doi:10.1016/j.jaci.2022.11.007. PMID   36543697. S2CID   254918066.
  19. 1 2 3 4 5 6 7 8 9 10 11 Ikeda T, Nishida A, Yamano M, Kimura I (November 2022). "Short-chain fatty acid receptors and gut microbiota as therapeutic targets in metabolic, immune, and neurological diseases". Pharmacology & Therapeutics. 239: 108273. doi: 10.1016/j.pharmthera.2022.108273 . PMID   36057320. S2CID   251992642.
  20. 1 2 Obradovic M, Sudar-Milovanovic E, Soskic S, Essack M, Arya S, Stewart AJ, Gojobori T, Isenovic ER (2021). "Leptin and Obesity: Role and Clinical Implication". Frontiers in Endocrinology. 12: 585887. doi: 10.3389/fendo.2021.585887 . PMC   8167040 . PMID   34084149.
  21. 1 2 Navalón-Monllor V, Soriano-Romaní L, Silva M, de Las Hazas ML, Hernando-Quintana N, Suárez Diéguez T, Esteve PM, Nieto JA (August 2023). "Microbiota dysbiosis caused by dietetic patterns as a promoter of Alzheimer's disease through metabolic syndrome mechanisms". Food & Function. 14 (16): 7317–7334. doi:10.1039/d3fo01257c. PMID   37470232. S2CID   259996464.
  22. 1 2 Shimizu H, Masujima Y, Ushiroda C, Mizushima R, Taira S, Ohue-Kitano R, Kimura I (November 2019). "Dietary short-chain fatty acid intake improves the hepatic metabolic condition via FFAR3". Scientific Reports. 9 (1): 16574. Bibcode:2019NatSR...916574S. doi:10.1038/s41598-019-53242-x. PMC   6851370 . PMID   31719611.
  23. 1 2 3 Pluznick JL, Protzko RJ, Gevorgyan H, Peterlin Z, Sipos A, Han J, Brunet I, Wan LX, Rey F, Wang T, Firestein SJ, Yanagisawa M, Gordon JI, Eichmann A, Peti-Peterdi J, Caplan MJ (March 2013). "Olfactory receptor responding to gut microbiota-derived signals plays a role in renin secretion and blood pressure regulation". Proceedings of the National Academy of Sciences of the United States of America. 110 (11): 4410–5. Bibcode:2013PNAS..110.4410P. doi: 10.1073/pnas.1215927110 . PMC   3600440 . PMID   23401498.
  24. 1 2 3 Ghislain J, Poitout V (March 2021). "Targeting lipid GPCRs to treat type2 diabetes mellitus - progress and challenges". Nature Reviews. Endocrinology. 17 (3): 162–175. doi:10.1038/s41574-020-00459-w. PMID   33495605. S2CID   231695737.
  25. 1 2 Yuan G, Wen S, Zhong X, Yang X, Xie L, Wu X, Li X (2023). "Inulin alleviates offspring asthma by altering maternal intestinal microbiome composition to increase short-chain fatty acids". PLOS ONE. 18 (4): e0283105. Bibcode:2023PLoSO..1883105Y. doi: 10.1371/journal.pone.0283105 . PMC   10072493 . PMID   37014871.
  26. Kim YA, Keogh JB, Clifton PM (June 2018). "Probiotics, prebiotics, synbiotics and insulin sensitivity". Nutrition Research Reviews. 31 (1): 35–51. doi: 10.1017/S095442241700018X . PMID   29037268.
  27. 1 2 Kimura I, Ichimura A, Ohue-Kitano R, Igarashi M (January 2020). "Free Fatty Acid Receptors in Health and Disease". Physiological Reviews. 100 (1): 171–210. doi: 10.1152/physrev.00041.2018 . PMID   31487233. S2CID   201845937.
  28. Loona DP, Das B, Kaur R, Kumar R, Yadav AK (2023). "Free Fatty Acid Receptors (FFARs): Emerging Therapeutic Targets for the Management of Diabetes Mellitus". Current Medicinal Chemistry. 30 (30): 3404–3440. doi:10.2174/0929867329666220927113614. PMID   36173072. S2CID   252598831.
  29. 1 2 3 Kuwahara A, Kuwahara Y, Inui T, Marunaka Y (March 2018). "Regulation of Ion Transport in the Intestine by Free Fatty Acid Receptor 2 and 3: Possible Involvement of the Diffuse Chemosensory System". International Journal of Molecular Sciences. 19 (3): 735. doi: 10.3390/ijms19030735 . PMC   5877596 . PMID   29510573.
  30. 1 2 3 4 5 Zhang D, Jian YP, Zhang YN, Li Y, Gu LT, Sun HH, Liu MD, Zhou HL, Wang YS, Xu ZX (August 2023). "Short-chain fatty acids in diseases". Cell Communication and Signaling. 21 (1): 212. doi: 10.1186/s12964-023-01219-9 . PMC   10436623 . PMID   37596634.
  31. Won YJ, Lu VB, Puhl HL, Ikeda SR (December 2013). "β-Hydroxybutyrate modulates N-type calcium channels in rat sympathetic neurons by acting as an agonist for the G-protein-coupled receptor FFA3". The Journal of Neuroscience. 33 (49): 19314–25. doi:10.1523/JNEUROSCI.3102-13.2013. PMC   3850046 . PMID   24305827.
  32. 1 2 Christiansen CB, Gabe MB, Svendsen B, Dragsted LO, Rosenkilde MM, Holst JJ (July 2018). "The impact of short-chain fatty acids on GLP-1 and PYY secretion from the isolated perfused rat colon". American Journal of Physiology. Gastrointestinal and Liver Physiology. 315 (1): G53–G65. doi: 10.1152/ajpgi.00346.2017 . PMID   29494208. S2CID   3633401.
  33. Mikami D, Kobayashi M, Uwada J, Yazawa T, Kamiyama K, Nishimori K, Nishikawa Y, Nishikawa S, Yokoi S, Taniguchi T, Iwano M (2020). "AR420626, a selective agonist of GPR41/FFA3, suppresses growth of hepatocellular carcinoma cells by inducing apoptosis via HDAC inhibition". Therapeutic Advances in Medical Oncology. 12: 1758835920913432. doi:10.1177/1758835920913432. PMC   7517987 . PMID   33014144.
  34. Hoyles L, Snelling T, Umlai UK, Nicholson JK, Carding SR, Glen RC, McArthur S (March 2018). "Microbiome-host systems interactions: protective effects of propionate upon the blood-brain barrier". Microbiome. 6 (1): 55. doi: 10.1186/s40168-018-0439-y . PMC   5863458 . PMID   29562936.
  35. 1 2 Teyani R, Moniri NH (August 2023). "Gut feelings in the islets: The role of the gut microbiome and the FFA2 and FFA3 receptors for short chain fatty acids on β-cell function and metabolic regulation". British Journal of Pharmacology. 180 (24): 3113–3129. doi:10.1111/bph.16225. PMID   37620991. S2CID   261121846.
  36. Al Mahri S, Malik SS, Al Ibrahim M, Haji E, Dairi G, Mohammad S (February 2022). "Free Fatty Acid Receptors (FFARs) in Adipose: Physiological Role and Therapeutic Outlook". Cells. 11 (4): 750. doi: 10.3390/cells11040750 . PMC   8870169 . PMID   35203397.
  37. Voltolini C, Battersby S, Etherington SL, Petraglia F, Norman JE, Jabbour HN (January 2012). "A novel antiinflammatory role for the short-chain fatty acids in human labor". Endocrinology. 153 (1): 395–403. doi: 10.1210/en.2011-1457 . hdl: 20.500.11820/7035e8fd-062a-491a-9943-f20ad03844dd . PMID   22186417. S2CID   34431654.
  38. Zamarbide M, Martinez-Pinilla E, Gil-Bea F, Yanagisawa M, Franco R, Perez-Mediavilla A (March 2022). "Genetic Inactivation of Free Fatty Acid Receptor 3 Impedes Behavioral Deficits and Pathological Hallmarks in the APPswe Alzheimer's Disease Mouse Model". International Journal of Molecular Sciences. 23 (7): 3533. doi: 10.3390/ijms23073533 . PMC   8999053 . PMID   35408893.
  39. 1 2 3 Kimura I, Inoue D, Maeda T, Hara T, Ichimura A, Miyauchi S, Kobayashi M, Hirasawa A, Tsujimoto G (May 2011). "Short-chain fatty acids and ketones directly regulate sympathetic nervous system via G protein-coupled receptor 41 (GPR41)". Proceedings of the National Academy of Sciences of the United States of America. 108 (19): 8030–5. Bibcode:2011PNAS..108.8030K. doi: 10.1073/pnas.1016088108 . PMC   3093469 . PMID   21518883.
  40. Cox MA, Jackson J, Stanton M, Rojas-Triana A, Bober L, Laverty M, Yang X, Zhu F, Liu J, Wang S, Monsma F, Vassileva G, Maguire M, Gustafson E, Bayne M, Chou CC, Lundell D, Jenh CH (November 2009). "Short-chain fatty acids act as antiinflammatory mediators by regulating prostaglandin E(2) and cytokines". World Journal of Gastroenterology. 15 (44): 5549–57. doi: 10.3748/wjg.15.5549 . PMC   2785057 . PMID   19938193.
  41. 1 2 3 Miyasato S, Iwata K, Mura R, Nakamura S, Yanagida K, Shindou H, Nagata Y, Kawahara M, Yamaguchi S, Aoki J, Inoue A, Nagamune T, Shimizu T, Nakamura M (January 2023). "Constitutively active GPR43 is crucial for proper leukocyte differentiation". FASEB Journal. 37 (1): e22676. doi: 10.1096/fj.202201591R . PMID   36468834. S2CID   254244683.
  42. Le Poul E, Loison C, Struyf S, Springael JY, Lannoy V, Decobecq ME, Brezillon S, Dupriez V, Vassart G, Van Damme J, Parmentier M, Detheux M (July 2003). "Functional characterization of human receptors for short chain fatty acids and their role in polymorphonuclear cell activation". The Journal of Biological Chemistry. 278 (28): 25481–9. doi: 10.1074/jbc.M301403200 . PMID   12711604.
  43. Yonezawa T, Kobayashi Y, Obara Y (January 2007). "Short-chain fatty acids induce acute phosphorylation of the p38 mitogen-activated protein kinase/heat shock protein 27 pathway via GPR43 in the MCF-7 human breast cancer cell line". Cellular Signalling. 19 (1): 185–93. doi:10.1016/j.cellsig.2006.06.004. PMID   16887331.
  44. Al Mahri S, Al Ghamdi A, Akiel M, Al Aujan M, Mohammad S, Aziz MA (May 2020). "Free fatty acids receptors 2 and 3 control cell proliferation by regulating cellular glucose uptake". World Journal of Gastrointestinal Oncology. 12 (5): 514–525. doi: 10.4251/wjgo.v12.i5.514 . PMC   7235185 . PMID   32461783.
  45. de Vos WM, Tilg H, Van Hul M, Cani PD (May 2022). "Gut microbiome and health: mechanistic insights". Gut. 71 (5): 1020–1032. doi:10.1136/gutjnl-2021-326789. PMC   8995832 . PMID   35105664.
  46. Iacobucci G (March 2023). "Appetite suppressant semaglutide is to be made available to treat obesity in England". BMJ (Clinical Research Ed.). 380: 556. doi:10.1136/bmj.p556. PMID   36889753. S2CID   257379156.
  47. Gabriel FC, Fantuzzi G (December 2019). "The association of short-chain fatty acids and leptin metabolism: a systematic review". Nutrition Research. 72: 18–35. doi:10.1016/j.nutres.2019.08.006. PMID   31669116. S2CID   155928278.
  48. Koh A, De Vadder F, Kovatcheva-Datchary P, Bäckhed F (June 2016). "From Dietary Fiber to Host Physiology: Short-Chain Fatty Acids as Key Bacterial Metabolites". Cell. 165 (6): 1332–1345. doi: 10.1016/j.cell.2016.05.041 . PMID   27259147. S2CID   8562345.
  49. Lee DH, Heo KS, Myung CS (July 2023). "Gαi -coupled GPR41 activation increases Ca2+ influx in C2C12 cells and shows a therapeutic effect in diabetic animals". Obesity. 31 (7): 1871–1883. doi:10.1002/oby.23786. PMID   37309717. S2CID   259147907.
  50. 1 2 Pluznick JL (April 2017). "Microbial Short-Chain Fatty Acids and Blood Pressure Regulation". Current Hypertension Reports. 19 (4): 25. doi:10.1007/s11906-017-0722-5. PMC   5584783 . PMID   28315048.
  51. 1 2 3 Kaye DM, Shihata WA, Jama HA, Tsyganov K, Ziemann M, Kiriazis H, Horlock D, Vijay A, Giam B, Vinh A, Johnson C, Fiedler A, Donner D, Snelson M, Coughlan MT, Phillips S, Du XJ, El-Osta A, Drummond G, Lambert GW, Spector TD, Valdes AM, Mackay CR, Marques FZ (April 2020). "Deficiency of Prebiotic Fiber and Insufficient Signaling Through Gut Metabolite-Sensing Receptors Leads to Cardiovascular Disease". Circulation. 141 (17): 1393–1403. doi: 10.1161/CIRCULATIONAHA.119.043081 . hdl: 10536/DRO/DU:30135388 . PMID   32093510. S2CID   211476145.
  52. 1 2 Dinakis E, Nakai M, Gill PA, Yiallourou S, Sata Y, Muir J, Carrington M, Head GA, Kaye DM, Marques FZ (November 2021). "The Gut Microbiota and Their Metabolites in Human Arterial Stiffness". Heart, Lung & Circulation. 30 (11): 1716–1725. doi:10.1016/j.hlc.2021.07.022. PMID   34452845. S2CID   237340692.
  53. Poll BG, Xu J, Jun S, Sanchez J, Zaidman NA, He X, Lester L, Berkowitz DE, Paolocci N, Gao WD, Pluznick JL (April 2021). "Acetate, a Short-Chain Fatty Acid, Acutely Lowers Heart Rate and Cardiac Contractility Along with Blood Pressure". The Journal of Pharmacology and Experimental Therapeutics. 377 (1): 39–50. doi:10.1124/jpet.120.000187. PMC   7985618 . PMID   33414131.
  54. 1 2 R Muralitharan R, Marques FZ (February 2021). "Diet-related gut microbial metabolites and sensing in hypertension". Journal of Human Hypertension. 35 (2): 162–169. doi:10.1038/s41371-020-0388-3. PMID   32733062. S2CID   220881080.
  55. Trompette A, Gollwitzer ES, Yadava K, Sichelstiel AK, Sprenger N, Ngom-Bru C, Blanchard C, Junt T, Nicod LP, Harris NL, Marsland BJ (February 2014). "Gut microbiota metabolism of dietary fiber influences allergic airway disease and hematopoiesis". Nature Medicine. 20 (2): 159–66. doi:10.1038/nm.3444. PMID   24390308. S2CID   35298402.
  56. 1 2 3 Ito T, Nakanishi Y, Shibata R, Sato N, Jinnohara T, Suzuki S, Suda W, Hattori M, Kimura I, Nakano T, Yamaide F, Shimojo N, Ohno H (2023). "The propionate-GPR41 axis in infancy protects from subsequent bronchial asthma onset". Gut Microbes. 15 (1): 2206507. doi:10.1080/19490976.2023.2206507. PMC   10158560 . PMID   37131293.
  57. Depner M, Taft DH, Kirjavainen PV, Kalanetra KM, Karvonen AM, Peschel S, Schmausser-Hechfellner E, Roduit C, Frei R, Lauener R, Divaret-Chauveau A, Dalphin JC, Riedler J, Roponen M, Kabesch M, Renz H, Pekkanen J, Farquharson FM, Louis P, Mills DA, von Mutius E, Ege MJ (November 2020). "Maturation of the gut microbiome during the first year of life contributes to the protective farm effect on childhood asthma". Nature Medicine. 26 (11): 1766–1775. doi:10.1038/s41591-020-1095-x. hdl: 2164/16359 . PMID   33139948. S2CID   226244474.
  58. Arrieta MC, Stiemsma LT, Dimitriu PA, Thorson L, Russell S, Yurist-Doutsch S, Kuzeljevic B, Gold MJ, Britton HM, Lefebvre DL, Subbarao P, Mandhane P, Becker A, McNagny KM, Sears MR, Kollmann T, Mohn WW, Turvey SE, Finlay BB (September 2015). "Early infancy microbial and metabolic alterations affect risk of childhood asthma". Science Translational Medicine. 7 (307): 307ra152. doi: 10.1126/scitranslmed.aab2271 . PMID   26424567. S2CID   206687974.