Targeting gut microbiota in bone health of livestock and poultry: roles of short-chain fatty acids

DOI: https://doi.org/10.21203/rs.3.rs-2019685/v1

Abstract

The regulation and maintenance of bone metabolic homeostasis plays a pivotal role in animal skeletal health. Several skeletal diseases have been confirmed to be closely related to ecological dysbiosis and structural changes in the gut microbiota. Gut microbiota and its metabolites, mainly short-chain fatty acids (SCFAs), affect almost all organs, including the skeleton. SCFAs positively affect bone healing by acting directly on cells involved in fracture healing or by shaping appropriate anti-inflammatory and immunomodulatory responses. Moreover, recent studies have shown that SCFAs play a biological role in regulating bone metabolism in four areas: immune function, calcium absorption, osteogenesis and osteolysis, thus havingpotential benefits in maintaining bone health in livestock and poultry. This review focuses on the role of SCFAs in the regulation of bone metabolism by gut microbiota and provides a theoretical basis for studies related to bone health in livestock and poultry.

Introduction

The bones form the skeleton, which serves multiple purposes, including support to the body’s weight, storing minerals like calcium and phosphorus, and producing blood cells in the bone marrow. Therefore, the regulation and maintenance of bone metabolic homeostasis plays a pivotal role in animal skeletal health. Bone metabolism is characterized by the close cooperation of bone cells (including osteoblasts, osteoclasts and osteoclasts) to maintain the number and integrity of bone microarchitecture, and once the homeostasis of bone metabolism is disrupted may lead to bone loss, consequently greatly increase the risk of skeletal diseases[1]. The normal processes of bone development and homeostasis are disrupted, leading to the development of bone disease, a phenomenon that is widespread throughout the modern poultry industry. Back in the early 1990s, leg disease caused an estimated $80–120 million per year in economic losses to the U.S. broiler industry. This issue still poses a greater threat to the broiler industry with the increased intensive management of poultry today[2], which not only leads to motor dysfunction accompanied by symptoms such as claudication, slow movement, and difficulty in standing[3], but also decreases animal performance and muscle quality[45], thereby resulting in huge economic losses. In addition, bone diseases are also common in animals such as pigs, cattle, and sheep. For example, long-term vitamin D deficiency predisposes to osteochondrosis, which leads to difficulty standing, lameness, and fractures[68].

Recent studies have found that gut microbiota is one of the important regulatory targets of bone homeostasis and bone health. Intestinal microbes can mediate bone homeostasis by participating in metabolic, immune and endocrine processes[2]. In the gastrointestinal tract (GIT), gut microbiota and host cells interact in ways that are normally beneficial to the host, including promoting the maturation of the intestinal immune system through interaction with immune cells (e.g., macrophages and dendritic cells); and maintaining the integrity of the intestinal barrier by inducing mucus production and providing nutrition to the intestinal epithelial cells[8]. However, alterations in the gut microbiota can disrupt the beneficial microbe-host relationship, which can lead to the development or progression of diseases, including inflammatory bowel disease, cardiovascular disease, asthma, and rheumatoid arthritis[911]. Several studies have confirmed that gut microbiota can affect bone metabolic homeostasis through different pathways and that short chain fatty acids (SCFAs), a metabolite of gut microbiota, play a key role in bone metabolism, including immunity, calcium absorption as well as deposition, osteogenesis and osteolysis[1214]. This paper reviews the current reports on the regulation of bone metabolic homeostasis by SCFAs, aiming to explore and introduce the regulatory role of SCFAs in bone metabolism and provide a theoretical basis for the prevention and treatment of skeletal diseases of livestock and poultry.

1. Gut Microbiota And Bone Metabolism

Bone metabolism occurs in a dynamic equilibrium between bone resorption and bone formation, and this process requires the synergistic action of both osteoclasts and osteoblasts[3]. Osteoblasts and osteoclasts play different roles in maintaining normal bone mass levels in animals. Osteoblasts are responsible for the synthesis, secretion and mineralization of bone matrix, whereas osteoclasts are mainly responsible for the resorption of the bone matrix[1516]. In addition, the synergistic effect of osteoblasts and osteoclasts is influenced by several factors, such as stem cell antigens, hormones and growth factors[17].

Animal tissues and organs are inextricably linked to each other. The intestine, as the largest immune organ in the animal body, plays an important role in the immune system and regulates the balance of bone metabolism[18]. Peek et al. confirmed that intestinal inflammation enhances the differentiation function of osteoclasts, significantly increasing bone loss and causing harm to bone health[19]. Moreover, the microorganism in the gut also has a key role in the regulation of bone mass. A study by Xi et al. demonstrated that favorable alterations in gut microecology under the influence of probiotics could alleviate bone loss caused by rheumatoid joints[20]. The above shows a strong correlation between gut and bone health.

In the GIT, microbes and its host have a complex mutually beneficial symbiotic relationship, resulting from their long-term co-evolution and mutual influence. These large and richly diverse microbial communities play an important role in the maintenance of bone health by regulating bone metabolism through their metabolites or gut microbiota-mediated regulators of bone metabolism. It has been shown that gut microbiota can regulate the ratio and relative activity of osteoclasts and osteoblasts through multiple pathways, thereby affecting bone metabolism and its normal growth and development[21]. SCFAs, metabolites of gut microbes, can regulate the differentiation, proliferation and apoptosis of osteoblasts through regulatory T cells (Tregs), thus affecting bone metabolic processes. In addition, Yan et al. reported that SCFAs could regulate serum insulin-like growth factor 1 levels and improve bone growth and health[22]. Therefore, SCFAs may be a key substance in regulating bone metabolism and preventing bone loss by the gut microbiota.

2. Scfas And Bone Health

2.1 Composition and origin of SCFAs

SCFAs, including a class of acid metabolites (C2-C5) such as acetic acid, propionic acid, butyric acid, valeric acid, isobutyl and isovaleric acid, are produced by bacteria in the gut of humans and animals by the fermentation of indigestible carbohydrates and proteins in food [23]. SCFAs are derived from the soluble dietary fiber found in foods such as oligosaccharides (bananas, Onions and asparagus), pectin (apples, apricots, carrots, oranges), kidney beans, oat bran, corn starch, milk, yogurt, and sprouted barley. Resistant starches such as barley, rice, beans, green bananas and potatoes are also important sources of SCFAs[2427]. These indigestible fibers are not digested and absorbed in the small intestine and are subsequently fermented by microbiota in the cecum and large intestine; SCFAs are mostly produced in the cecum aswell asproximal colon and less in the distal colon[28]. Among them, acetic acid, propionic acid and butyric acid account for 90%−95% of the total amount of SCFAs produced by gut microbiota and are the main components of intestinal SCFAs[29]. Therefore, the above-mentioned three kinds of SCFAs have been studied extensively, especially butyric acid.

2.2 Biological functions of SCFAs

SCFAs are closely related to intestinal microecology and are involved in a wide-range of biological processes, including signal transduction, regulation of cytokines, immune cells and intestinal mucosal barrier function[3031]. Furthermore, SCFAs increase bone formation and improve bone quality through regulating immunity, intestinal barrier function and immune cell activity.

SCFAs can be directly transported throughout the body by the host cells at the intestinal endothelial barrier, thus affecting distant tissues. G protein-coupled receptors (GPRs) are capable of binding to SCFAs, including GPR43, GPR41 and GPR109a[32]. These membrane-bound receptors are expressed on various immune cell types, including monocyte macrophages, and on other non-immune cells such as intestinal epithelial cells, adipocytes and enteroendocrine cells[33]. When these receptors bind to SCFA, it leads to intracellular Ca2+ release and activation of different downstream signaling pathways, such as ERK/MAPK, p38 or Akt/PI3K signaling cascades, thus regulating cellular activity and function[34]. In addition, some SCFAs, such as butyrate, serve as the main source of energy for the vital activities of intestinal epithelial cells, which directly affect the growth and development of these cells. Nonetheless, a certain number of SCFAs reach the bloodstream through transport systems (SMCT1/Slc5a8 pathway, MCT1/Slc16a1 pathway or passive diffusion) and are transported to the whole body through the transport system. Once SCFAs enter the circulatory system, they will affect the metabolism and function of peripheral tissues (adipose tissue, skeletal muscle, bone, etc.) by activating GPRs[3536].

2.2.1 Butyric acid (C4) and bone health

However, butyric acid exhibits the most extensive biological activity among SCFAs, including regulating inflammation, maintaining immune homeostasis, and reducing bone loss from inflammation. Butyric acid can downregulate the pro-inflammatory mediators NO, IL-6 and IL-12 produced by lipopolysaccharide (LPS)-induced macrophages and can also promote the production of IL-22 through GPR41 and HDAC inhibition[37]. Butyric acid inhibits LPS-induced maturation and biological activity of monocyte-derived dendritic cells and promotes the polarization of early CD4 + T cells into IL-10-producing Tregs[38]. While, butyric acid regulates the inflammatory state of the body by activating GPRs in the intestinal mucosal epithelium, reducing the synthesis and secretion of pro-inflammatory factors such as Tumor necrosis factor-α(TNF-α) and Cyclooxygenase-2, thereby reducing the bone loss brought about by inflammation[10, 39].

Furthermore, butyric acid alleviates intestinal inflammation and reduces osteoclast differentiation by attenuating TNF-α-mediated immune responses and reducing inflammatory vesicles such as NLRP3[40]. Butyrate has also been reported to regulate Claudin-2 expression, reduce intestinal permeability through an IL-10 receptor-dependent mechanism, and strengthen intestinal barrier function by increasing colonic mucin and tight junction protein production[1, 41], enhancing immune response system function thus reducing bone loss[42]. Kaisar et al. found that butyrate inhibited the expression of the pro-inflammatory factor IFN-γ caused by the overactivation of the IFN-γ/ STAT1 signaling pathway[43]. Moreover, butyrate can also modulate the immune effect and relieve osteoarthritis by inhibiting the activity of inflammation-related pathways NF-κB and JAK/ STAT and IL-12p70, IL-23, and preventing the polarization of early CD4+ T lymphocytes into Th1(T helper cells 1) and Th17[44]. In short, Butyric acid has a positive effect on the maintenance of bone health by regulating the immune function of the animal body and alleviating bone destruction and loss.

2.2.2 Acetic acid (C2) and bone health

Acetic acid is one of the most abundant SCFAs produced by the gut microbiota[45]. It positively impacts bone health by maintaining the integrity of the intestinal mucosal barrier, preventing the invasion of pathogenic bacteria and enhancing the host's immune function. In animals, acetic acid is mainly present as free acid in tissues, excreta and blood[46]. It was found that acetic acid increases the production of IgA in the colon, alters the ability of IgA to bind to specific intestinal bacteria, and alters the colonization of these bacteria, enhancing the immune barrier function of the intestinal mucosa, thereby indirectly reducing the release of pro-inflammatory factors (TNF-α, IL-1β, etc.), inhibiting osteoclast activity and reducing bone loss[47, 48]. In addition, acetic acid can activate the GPR41 receptor on the surface of immune cells, enhancing the immune effect and facilitating the maintenance of bone health[4950]. Maslowski et al. displayed that acetic acid significantly improved intestinal function, reduced DNA-dependent activator of IFN-regulatory factors and inflammatory mediator myeloperoxidase levels and TNF-α, thus facilitating the remission of the inflammatory response and reducing osteoclast production and differentiation[51]. The above facts indicated that acetic acid enhances immune function and reduces the release of pro-inflammatory factors, thereby inhibiting the activation of osteolytic effects.

2.2.3 Propionic acid (C3) and bone health

Propionic acid is an organic acid that occurs naturally as a result of the kind of bacterial action found on the skin or in the GIT. Propionic acid in the gut plays a variety of roles after passing through the bloodstream, including affecting hepatic cholesterol metabolism, promoting calcium absorption, increasing calcium deposition, and facilitating bone production[5253]. In addition, it was found that propionic acid not only activates NLRP3 inflammatory vesicles in intestinal epithelial cells, induces IL-18 secretion, and improves the integrity of the intestinal mucosal epithelial barrier, but also inhibits histone deacetylase and decreases NF-κB activity, thereby reducing the release of inflammatory factors TNF-α, IL-6, and IL-8 and affecting the structure and function of the intestinal mucosal barrier[54]. Osteoporosis is closely associated with the cellular imbalance of the immune system and immune-mediated effects on bone structure through the intestine have been demonstrated[55]. Interestingly, SCFAs intake resulted in increased bone mass in mice accompanied by a decrease in inflammation-induced bone loss. In a study of the effects of propionic acid supplementation on human bone metabolism, Duscha et al. found that propionic acid intake induced a significant increase in serum osteocalcin (a marker of bone formation) levels and a significant decrease in β-CrossLaps (a marker of bone resorption) levels; suggesting that propionic acid induces increased bone formation and decreased bone resorption[56].

2.2.4 Valeric acid (C5) and bone health

The GIT displayed high propionate and butyrate levels and low valeric acid levels[57]. However, Yuille's study confirmed that total specific inhibition of HDAC and promotion of differentiation of Tregs into T cells by increasing the ability of intestinal flora to produce butyric and valeric acid could enhance bone immunity indirectly[58]. Studies have shown that although valeric acid levels are low in animals, dietary fiber intake helps to increase valeric acid levels and that higher valeric acid levels reduce the release of pro-inflammatory factors and mitigate bone destruction[58, 59]; suggesting that the immunomodulatory ability of valeric acid and its potential therapeutic value for inflammation-induced bone diseases.

There are similarities in the functions of the different SCFAs. For example, they can regulate the structure of the gut microbiota (regulating pH in the gut and reducing the colonization of pathogenic bacteria), which facilitates the establishment of the intestinal immune barrier, and inhibit the release of inflammation-related signaling molecules (IL-6, IL-7, Receptor activator for nuclear factor-κB ligand, (RANKL)), which reduces osteoclast differentiation and thus facilitates bone health. The above facts show that intestinal health is closely related to bone health.

3. Gut Microbiota And Scfas

There are differences in the type of SCFAs produced by fermentation of different kinds of gut microbiota (As shown in Table 1). According to Wolin et al., the fermentation products of Lactobacillus spp. are mainly lactic acids. The fermentation products of Synechococcus spp. are mainly acetic acid and butyric acid, whereas the fermentation products of Bifidobacterium spp. are mostly acetic acid, lactic acid, and formic acid[60]. It can be seen that different gut microbiota produce different SCFAs. In addition, dietary fiber is essential in terms of the composition and metabolic activity of the gut microbiota and can influence the level of SCFAs. Diet has a decisive role in the structure of the gut microbiota and the level of SCFAs. Therefore, it is possible to modify the structure of gut microbiota through diet to regulate SCFAs. Even short-term dietary interventions can have a significant impact on gut microbiota structure. In particular, those diets based exclusively on animal products, consuming reduced-fat foods high in protein and low in carbohydrates or fiber, can increase the relative abundance of the Bacteroidetes and decrease the relative quantity of the Firmicutes, leading to an imbalance in the structure of the microorganism, while affecting SCFAs concentrations in the intestine[61]. In conclusion, long-term poor dietary habits may increase the risk of intestinal diseases, and the intake of more fermentable dietary fiber can regulate the structure of the microorganism and thus positively affect the prevention of diseases.

When studying the concentration of SCFAs under different gut microbiota structures, it was found that the ability of the gut microbiota to produce SCFAs was enhanced by adding certain probiotics[62]. For example, in a broiler cecum model, supplementation with Lsalivarius ssp. increased the concentration of propionate and butyrate in the cecum[63]. Furthermore, in previous studies by Guss, it was found that changing the composition of gut microbiota by taking antibiotics, dietary changes and adding probiotics can affect bone health[64]. These studies indicate that the structure of gut microbiota can be altered by diet, which further leads to changes in SCFA concentration, and that different species of gut microbiota produce different types and amounts of SCFAs. In addition, it also suggests that the gut microbiota may have a regulatory effect on bone metabolism through SCFAs.

4. Regulation Of Scfas On Bone Metabolism

As bioinformatics and molecular biotechnology have developed, increasing studies have demonstrated gut microbiota metabolites regulate bone metabolism[6566]. Among its metabolites, SCFAs have been shown to affect almost all body organs, including the bone[67]. SCFAs can participate in bone metabolism by directly acting on osteoblasts, osteoclasts, chondrocytes, and fibroblasts, or indirectly by regulating the absorption of mineral elements, and can also affect bone metabolism by modulating the immune system[6869]. SCFAs exhibit a wide range of bone metabolism activities and positively affect bone quality improvement.

4.1 SCFAs regulate bone metabolism through the immune system

SCFAs promote the maturation of the host immune system and regulate bone metabolism through the immune system[70]. Studies have shown that gut microbiota can interact with immune cells and dendritic cells to promote the production of molecules such as SCFAs, indole derivatives, polyamines and secondary bile acids[71]. Among them, the receptors of SCFAs are expressed on immune cells, and the binding of SCFA to the corresponding receptors has a regulatory effect on T-cell function and immune cell differentiation, thereby enhancing immune function and reducing bone loss caused by inflammation. In addition, there is evidence that in osteoporosis caused by estrogen deficiency, T cells can increase the production of pro-inflammatory and pro-osteoclastic cytokines in bone tissue, such as TNF-α and RANKL, and that the upregulation of the expression of these factors in osteoblasts enhances the osteoclastogenesis induced by Th17 to stimulate the regulation of bone resorption[72]. SCFAs play an important role in bone metabolism and bone mineral density due to their close relationship with immune cells and bone cells.

In addition, it has been reported that butyrate and propionate can regulate intestinal immune function by inhibiting histone deacetylase (HDAC)[73]. Chang et al. demonstrated that butyrate produced by gut microbiota acts as an inhibitor of HDAC and modulates the function of macrophages in the lamina propria of the mouse gut[74]. Related studies have shown that the inhibition of HDAC increases the development and function of Tregs. Therefore, it may be one of the mechanisms by which SCFAs enhance the production of Tregs in the GIT[75]. The interaction of SCFAs with GPRs not only drives the differentiation of T cells into Tregs, but also promotes differentiation into effector T cells[76]. Park and colleagues suggested that SCFAs may induce helper T cells to differentiate into Th1 and Th17, thereby increasing host resistance to pathogen attack[7778]. SCFAs such as butyrate and propionate also regulate antigen presentation through HDAC inhibition by interacting with GPRs and have an inhibitory effect on dendritic cell development. Together, it can be seen that gut microbiota can affect the overall immune function of the host through SCFAs, immune cells and immune factors act on bone tissue, to affect the biochemical activity of osteoblasts in this process, and exert a regulatory effect on bone homeostasis and bone mass.

4.2 SCFAs affect calcium absorption and increase BMD and bone mechanical strength

SCFAs can increase calcium element deposition and bone mineral density (BMD) and mechanical strength. Calcium is an essential element for bone formation and sufficient calcium is necessary to improve bone quality and increase bone mechanical strength[7980]. Maintaining calcium balance plays a crucial role in achieving good peak bone mass and inhibiting the development of bone loss. Prebiotics are food components that selectively stimulate the growth or activity of one or several bacteria in the colon, thus exerting a beneficial effect on the host and having a role in regulating the gut microbiota structure [81]. It has been reported that SCFAs produced by the fermentation of prebiotics by gut microbiota can promote calcium absorption and significantly improve bone strength and bone mineral density in mice[82]. A study found that daily prebiotic fiber intake can enhance calcium absorption in adolescent children, which is beneficial to the bone development of adolescent-aged children[83]. Dietary fiber intake affects calcium absorption: increased levels of intestinal SCFAs after fermentation by gut microbiota can reduce the pH of the intestinal microecology, thus reducing the formation of calcium phosphate and increasing calcium production and absorption[84]. A study by Wallimann et al. confirmed that butyric acid significantly increases calcium deposition at the site of bone injury and promotes bone healing[85].

Furthermore, the effect of SCFAs on calcium deposition may not only be reflected in changes in intestinal pH. Indeed, SCFAs have been shown to increase calcium transport by regulation of signaling pathways[86, 87]. Furthermore, the SCFAs can indirectly improve calcium absorption by modulating the production of intestinal serotonin, namely 5-hydroxytryptamine (5-HT)[88]. Serotonin, a molecule that interacts with osteoblasts, has been used as a potential regulator of bone mass to prevent osteoporosis by increasing bone formation[89]. Duodenal enterochromaffin cells have the biological function of synthesizing 5-HT, and can promote the synthesis of 5-HT under the action of SCFAs[90]. It has been reported that 5-HT can interact with osteoblasts, especially by activating the 5-HT1B receptor on preosteoblasts to reduce osteoblast proliferation and thus improve the bone loss caused by osteoporosis[91]. In conclusion, SCFAs can directly or indirectly regulate bone formation, thereby increasing bone mineral density and bone strength and reducing fracture risk.

4.3 The regulatory role of SCFAs in osteoblasts

SCFAs can promote bone formation by regulating osteoblast activity. Osteoblasts have a pivotal role in bone formation and activation of Wnt signaling in osteoblasts is critical for osteoblast proliferation and bone homeostasis[87]. Studies have demonstrated that the Wnt signaling pathway has a crucial role in bone development and endostasis; moreover, SCFAs can activate the wnt pathway and induce the expression of the transcription factor osterix, thereby promoting osteoblast differentiation[49, 87]. In addition, this signaling pathway induces the expression of osteoprotegerin (OPG), an osteoclast suppressor in osteoblast lineage cells, inhibiting bone resorption[2]. It was found that butyrate can affect normal osteoblasts, increase osteoblast mineralization to promote bone formation and inhibit osteoclast differentiation by promoting OPG production in a human study[92].

SCFAs can indirectly regulate bone metabolism through Tregs. Tregs are cells that regulate the body’s immune function by actively regulating the activation and proliferation of potentially self-reactive T cells present in the normal organism. In addition to their immunomodulatory functions, Tregs can also exert some regulatory effects on bone homeostasis. They inhibit osteoclastogenesis, promote osteoblast differentiation, and are required for parathyroid hormone-stimulated bone formation[93]. Tregs were isolated from SPF mice and cultured in vitro with SCFAs. It was found that in the presence of propionate, the proliferation of Tregs can be promoted, thereby enhancing the regulation of bone homeostasis by Tregs[94]. Supplementation with probiotics can alleviate pathological bone loss to some extent. Our study has demonstrated that L. rhamnosus JYLR-005 prevented thiram-induced Tibial Dyschondroplasia by improving bone-related growth performance in broilers, including tibia weight, length, and mean diameter[95]. Tyagi et al. found that supplementation of L. rhamnosus in mouse diets can affect bone homeostasis, and the results presented that L. rhamnosus increased the volume of bone trabeculae and promoted increased bone formation[96]. It was attributed to the production of butyrate in the gut after L. rhamnosus ingestion, which induced the proliferation of Tregs in the intestine and bone tissue. The same results were obtained in an experiment where butyrate was fed directly to germ-free mice[97]. These studies suggested that SCFAs may indirectly regulate bone homeostasis through the biological function of Tregs.

4.4 The regulatory role of SCFAs in osteoclasts

SCFAs can modulate the activity of osteoclasts, which in turn regulates bone resorption. Osteoclasts are key cells involved in the regulation of bone resorption and are essential for maintaining the homeostasis of bone metabolism. Montalvany-Antonucci studied the effect of SCFA on alveolar bone and found that SCFA acts as a regulator of bone resorption and reduces osteoclast differentiation dependent on the activation of free fatty acid receptor 2[98].

Osteoclast generation and bone resorption are energy-consuming processes closely related to energy metabolism. Lemma demonstrated that the energy required for osteoclast differentiation mainly from oxidative phosphorylation, while peripheral cellular activities associated with bone matrix degradation are powered by glycolysis[99]. In the study of Lucas et al., the protective effect of SCFAs on bone mass was associated with inhibition of osteoclast differentiation and bone resorption. It is because propionate and butyrate induce a shift in the metabolic direction of osteoclasts, leading to enhanced glycolysis without significant changes in oxidative phosphorylation levels, resulting in downregulation of essential osteoclast genes, such as TRAF6 and NFATc1, affecting RANKL-induced osteoclast differentiation, thereby reducing the number of osteoclasts and regulating bone homeostasis[100]. Wauquier et al. found that GPR40 receptor-deficient mice exhibited features of osteoporosis, suggesting that GPR40 receptors positively affect bone density and demonstrating a mediating role for GPR40 receptors in fatty acid-induced bone remodeling[101]. An in vitro experimental study by Wallimann et al. revealed that many genes related to osteoclast differentiation after butyric acid treatment were differentially present in osteoclast precursor cells. Moreover, differentially expressed genes on osteoblast precursor cells can markedly reduce osteoclast formation and bone resorption activity[85]. Based on the above findings, SCFAs are shown to be an effective regulator of osteoclast metabolism and bone homeostasis.

5. Conclusion And Prospects

In recent years, the problem of bone diseases has become more prevalent as livestock and poultry industries have become increasingly intensive. Effective control and treatment of skeletal diseases is an urgent need of era, and it is especially important to find reasonable methods and approaches for prevention and treatment. There has been continuous evidence that SCFAs that act indirectly on gut microbiota regulation can modulate bone metabolism and reduce bone loss directly, thereby positively contributing to bone health. SCFAs modulate the immune system, calcium absorption, and bone cell regulation, thereby contributing to bone health (Table 2). Therefore, providing animals with appropriate SCFAs can improve bone health and their growth performance. The complex effects of SCFAs on skeletal development offer the possibility of more efficient treatment of bone diseases. However, More studies are required to determine the optimal ratios and doses for clinical application, and the effects of SCFAs on skeletal development. In the future, with the development of biomedical technology, we anticipate that the potential role of SCFAs in skeletal development will be further explored; SCFAs are crucial for the regulation of bone metabolism and maintenance of bone health, and these new findings will make clinical treatment of bone diseases even more effective, better serve livestock and poultry producers.

Abbreviations

BMD, Bone mineral density; GIT, Gastrointestinal tract; GPRs, G protein-coupled receptor; HDAC, Histone deacetylase; 5-HT, 5-hydroxytryptamine; LPS, Lipopolysaccharide; OPG, Osteoprotegerin; RANKL, Receptor activator for nuclear factor-κB ligand; SCFAs, Short chain fatty acids; Th, T helper cells; TNF-α, Tumor necrosis factor-α; Tregs, regulatory T cells.

Declarations

Author contributions

C.H contributed to the conceptualization, resources, funding acquisition, and revision and editing of the manuscript. Y.F.H. performed the literature review and drafted the manuscript. A.S. contributed to the revision and editing of the manuscript. L.X.L. and T.T.X. collected the literature and reviewed the text. All authors contributed to the article and approved the submitted version.

Funding

This study was supported by the China Postdoctoral Science Foundation (No. 2020M672234), the Outstanding Talents of Henan Agricultural University (No.30500421), and the Key Scientific Research Project of Henan Higher Education Institutions of China (No. 21A230013).

Conflict of Interest:

The authors declare no competing or financial interests.

References

  1. Barsony J, Kleess L, Verbalis JG (2019) Hyponatremia Is Linked to Bone Loss, Osteoporosis, Fragility and Bone Fractures. Front Horm Res 52:49–60. https://doi.org/10.1159/000493237
  2. Xu T, Yue K, Zhang C, Tong X, Lin L, Cao Q, Huang S (2022) Probiotics Treatment of Leg Diseases in Broiler Chickens: a Review. Probiotics Antimicrob Proteins 14(3):415–425. https://doi.org/10.1007/s12602-021-09869-2
  3. Xu T, Zheng J, Jin W, Li L, Lin L, Shaukat A, Zhang C, Cao Q, Ashraf M, Huang S (2022) Total Flavonoids of Rhizoma Drynariae Ameliorate Bone Growth in Experimentally Induced Tibial Dyschondroplasia in Chickens via Regulation of OPG/RANKL Axis. Frontiers in pharmacology 13:881057. https://doi.org/10.3389/fphar.2022.881057
  4. Huang SC, Cao QQ, Cao YB, Yang YR, Xu TT, Yue K, Liu F, Tong ZX, Wang XB (2021) Morinda officinalis polysaccharides improve meat quality by reducing oxidative damage in chickens suffering from tibial dyschondroplasia. Food Chem 344:128688. https://doi.org/10.1016/j.foodchem.2020.128688
  5. Cao QQ Kong AA, Tao KS, Zheng SH, Tong C, Wang XB, Tong ZX, Rehman MU, Huang SC (2020) Characterization of growth performance, meat quality and serum biochemical parameters in chickens suffering from tibial dyschondroplasia. Livestock Science 233:103956. https://doi.org/10.3389/fphys.2022.887207
  6. Hejazi R, Danyluk AJ (2009) Epidemiological investigation of femoral fractures in market pigs and the associated economic implications. Can Vet J 50(5):516–518. https://doi.org/10.2376/0005-9366-122-219
  7. Yadav GP, Sangwan V, Kumar A (2019) Comparative occurrence pattern of fractures in cattle and buffaloes. Vet World 12(7):1154–1159. https://doi.org/10.14202/vetworld.2019.1154-1159
  8. Tóth F, David FH, LaFond E, Wang L, Ellermann JM, Carlson CS (2017) In vivo visualization using MRI T2 mapping of induced osteochondrosis and osteochondritis dissecans lesions in goats undergoing controlled exercise. J Orthop Res 35(4):868–875. https://doi.org/10.1002/jor.23332
  9. Xu T, Yue K, Zhang C, Tong X, Lin L, Cao Q, Huang S (2022) Probiotics Treatment of Leg Diseases in Broiler Chickens: a Review. Probiotics Antimicrob Proteins 14(3):415–425. https://doi.org/10.1007/s12602-021-09869-2
  10. Zhang Z, Zhang H, Chen T, Shi L, Wang D, Tang D (2022) Regulatory role of short-chain fatty acids in inflammatory bowel disease. Cell Commun Signal 20(1):64. https://doi.org/10.1186/s12964-022-00869-5
  11. Deal C (2012) Bone loss in rheumatoid arthritis: systemic, periarticular, and focal. Curr Rheumatol Rep. 14(3):231–7. https://doi.org/10.1007/s11926-012-0253-7
  12. Zhang Z, Lin T, Meng Y, Hu M, Shu L, Jiang H, Gao R, Ma J, Wang C, Zhou X (2021) FOS/GOS attenuates high-fat diet induced bone loss via reversing microbiota dysbiosis, high intestinal permeability and systemic inflammation in mice. Metabolism 119:154767. https://doi.org/10.1016/j.metabol.2021.154767
  13. Holscher HD (2017) Dietary fiber and prebiotics and the gastrointestinal microbiota. Gut Microbes. 8(2):172–184. https://doi.org/10.1080/19490976.2017.1290756
  14. Sanders ME, Merenstein DJ, Reid G, Gibson GR, Rastall RA (2019) Probiotics and prebiotics in intestinal health and disease: from biology to the clinic. Nat Rev Gastroenterol Hepatol 16(10):605–616. https://doi.org/10.1038/s41575-019-0173-3
  15. Hu K, Olsen BR (2016) Osteoblast-derived VEGF regulates osteoblast differentiation and bone formation during bone repair. J Clin Invest. 126(2):509–26. https://doi.org/10.1172/JCI82585
  16. Ono T, Nakashima T (2018) Recent advances in osteoclast biology. Histochem Cell Biol 149(4):325–341. https://doi.org/10.1007/s00418-018-1636-2
  17. Chen X, Wang Z, Duan N, Zhu G, Schwarz EM, Xie C (2018) Osteoblast-osteoclast interactions. Connect Tissue Res 59(2):99–107. https://doi.org/10.1080/03008207.2017.1290085
  18. Alonso C, Vicario M, Pigrau M, Lobo B, Santos J (2014) Intestinal barrier function and the brain-gut axis. Adv Exp Med Biol 817:73–113. https://doi.org/10.1007/978-1-4939-0897-4_4
  19. Peek CT, Ford CA, Eichelberger KR et al (2022) Intestinal inflammation promotes MDL-1+ osteoclast precursor expansion to trigger osteoclastogenesis and bone loss. Cell Mol Gastroenterol Hepatol. S2352-345X(22)00160-6. https://doi.org/10.1016/j.jcmgh.2022.07.002
  20. Xi X, Ye Q, Li X, Lu X, Fan D, Xia Y, Xiao C (2022) Xiong Fu Powder Regulates the Intestinal Microenvironment to Protect Bones Against Destruction in Collagen-Induced Arthritis Rat Models. Front Cell Infect Microbiol 12:854940. https://doi.org/10.3389/fcimb.2022.854940
  21. Behera J, Ison J, Tyagi SC, Tyagi N (2020) The role of gut microbiota in bone homeostasis. Bone 135:115317. https://doi.org/10.1302/2046-3758.101.BJR-2020-0273.R1
  22. Yan J, Herzog JW, Tsang K, Brennan CA, Bower MA, Garrett WS, Sartor BR, Aliprantis AO, Charles JF (2016) Gut microbiota induce IGF-1 and promote bone formation and growth. Proc Natl Acad Sci U S A 113(47):E7554-E7563. https://doi.org/10.1073/pnas.1607235113
  23. Sam QH., Ling H, Yew WS, Tan Z, Ravikumar S, Chang MW, Chai L (2021) The Divergent Immunomodulatory Effects of Short Chain Fatty Acids and Medium Chain Fatty Acids. Int J Mol Sci 22(12):6453. https://doi.org/10.3390/ijms22126453
  24. P NPV, Joye IJ (2020) Dietary Fibre from Whole Grains and Their Benefits on Metabolic Health. Nutrients 12(10):3045. https://doi.org/10.3390/nu12103045
  25. Barber TM, Kabisch S, Pfeiffer AFH, Weickert MO (2020) The Health Benefits of Dietary Fibre. Nutrients 12(10):3209. https://doi.org/10.3390/nu12103209
  26. Cronin P, Joyce SA, O'Toole PW, O'Connor EM (2021) Dietary Fibre Modulates the Gut Microbiota. Nutrients 13(5):1655. https://doi.org/10.3390/nu13051655
  27. Li YJ, Chen X, Kwan TK et al (2020) Dietary Fiber Protects against Diabetic Nephropathy through Short-Chain Fatty Acid-Mediated Activation of G Protein-Coupled Receptors GPR43 and GPR109A. J Am Soc Nephrol 31(6):1267–1281. https://doi.org/10.1681/ASN.2019101029
  28. Sun M, Wu W, Liu Z, Cong Y (2017) Microbiota metabolite short chain fatty acids, GPCR, and inflammatory bowel diseases. J Gastroenterol 52(1):1–8. https://doi.org/10.1007/s00535-016-1242-9
  29. Wong JM, de Souza R, Kendall CW, Emam A, Jenkins DJ (2006) Colonic health: fermentation and short chain fatty acids. J Clin Gastroenterol 40(3):235–43. https://doi.org/10.1097/00004836-200603000-00015
  30. Louis P, Flint HJ (2017) Formation of propionate and butyrate by the human colonic microbiota. Environ Microbiol 19(1): 29–41. https://doi.org/10.1111/1462-2920.13589
  31. Tan J, McKenzie C, Potamitis M, Thorburn AN, Mackay CR, Macia L (2014) The role of short-chain fatty acids in health and disease. Adv Immunol 121:91–119. https://doi.org/10.1016/B978-0-12-800100-4.00003-9
  32. Zaiss MM, Jones RM, Schett G, Pacifici R (2019) The gut-bone axis: how bacterial metabolites bridge the distance. J Clin Invest 129(8):3018–3028. https://doi.org/10.1172/JCI128521
  33. Sun G, Wang Y, Ti Y, Wang J, Zhao J, Qian H (2017) Regulatory B cell is critical in bone union process through suppressing proinflammatory cytokines and stimulating Foxp3 in Treg cells. Clin Exp Pharmacol Physiol 44(4):455–462. https://doi.org/10.1111/1440-1681.12719
  34. Melhem H, Kaya B, Ayata CK, Hruz P, Niess JH (2019) Metabolite-Sensing G Protein-Coupled Receptors Connect the Diet-Microbiota-Metabolites Axis to Inflammatory Bowel Disease. Cells 8(5):450. https://doi.org/10.3390/cells8050450
  35. Li G, Lin J, Zhang C, Gao H, Lu H, Gao X, Zhu R, Li Z, Li M, Liu Z (2021) Microbiota metabolite butyrate constrains neutrophil functions and ameliorates mucosal inflammation in inflammatory bowel disease. Gut microbes 13(1):1968257. https://doi.org/10.1080/19490976.2021.1968257
  36. Zhou D, Fan JG (2019) Microbial metabolites in non-alcoholic fatty liver disease. World J Gastroenterol 25(17):2019–2028. https://doi.org/10.3748/wjg.v25.i17.2019
  37. Nastasi C, Fredholm S, Willerslev-Olsen A et al (2017) Butyrate and propionate inhibit antigen-specific CD8+ T cell activation by suppressing IL-12 production by antigen-presenting cells. Sci Rep 7(1):14516. https://doi.org/10.1038/s41598-017-15099-w
  38. Park JH, Kotani T, Konno T, Setiawan J, Kitamura Y, Imada S, Usui Y, Hatano N, Shinohara M, Saito Y, Murata Y, Matozaki T (2016) Promotion of intestinal epithelial cell turnover by commensal bacteria: role of short-chain fatty acids. PLoS One 11(5):e0156334. https://doi.org/10.1371/journal.pone.0156334
  39. Yang Q, Ouyang J, Sun F, Yang J (2020) Short-chain fatty acids: a soldier fighting against inflammation and protecting from tumorigenesis in people with diabetes. Front Immunol 11:590685. https://doi.org/10.3389/fimmu.2020.590685
  40. Clark A, Mach N (2017) The crosstalk between the gut microbiota and mitochondria during exercise. Front Physiol 8:319. https://doi.org/10.3389/fphys.2017.00319
  41. Gonzalez A, Krie R, Massey HD, Carl D, Ghosh S, Gehr T, Ghosh, SS (2019) Sodium butyrate ameliorates insulin resistance and renal failure in CKD rats by modulating intestinal permeability and mucin expression. Nephrol Dial Transplant 34(5):783–794. https://doi.org/10.1093/ndt/gfy238
  42. Yan H, Ajuwon KM (2017) Butyrate modifies intestinal barrier function in IPEC-J2 cells through a selective upregulation of tight junction proteins and activation of the Akt signaling pathway. PLoS One 12(6):e0179586. https://doi.org/10.1371/journal.pone.0179586
  43. Kaisar M, Pelgrom LR, van der Ham AJ, Yazdanbakhsh M, Everts B (2017) Butyrate conditions human dendritic cells to prime type 1 Tregs via both histone deacetylase inhibition and G protein-coupled receptor 109A signaling. Front Immunol 8:1429. https://doi.org/10.3389/fimmu.2017.01429
  44. Dalile B, Van Oudenhove L, Vervliet B, Verbeke K (2019) The role of shortchain fatty acids in microbiota-gut-brain communication. Nat Rev Gastroenterol Hepatol 16(8):461–478. https://doi.org/10.1038/s41575-019-0157-3
  45. Lavelle A, Sokol H (2020) Gut microbiota-derived metabolites as key actors in inflammatory bowel disease. Nat Rev Gastroenterol Hepatol 17:223–237. https://doi.org/10.1038/s41575-019-0258-z
  46. Deleu S, Machiels K, Raes J, Verbeke K, Vermeire S (2021) Short chain fatty acids and its producing organisms: an overlooked therapy for IBD? EBioMedicine 66:103293. https://doi.org/10.1016/j.ebiom.2021.103293
  47. Boets E, Gomand SV, Deroover L et al (2017) Systemic availability and metabolism of colonic-derived short-chain fatty acids in healthy subjects: a stable isotope study. J Physiol 595:541–555. https://doi.org/10.1113/JP272613
  48. Takeuchi T, Miyauchi E, Kanaya T et al (2021) Acetate differentially regulates IgA reactivity to commensal bacteria. Nature 595(7868):560–564. https://doi.org/10.1038/s41586-021-03727-5
  49. Kobayashi Y, Uehara S, Udagawa N, Takahashi N (2016) Regulation of bone metabolism by Wnt signals. J Biochem 159(4):387–392. https://doi.org/10.1093/jb/mvv124
  50. Le Poul E, Loison C, Struyf S et al (2003) Functional characterization of human receptors for short chain fatty acids and their role in polymorphonuclear cell activation. J Biol Chem 278(28):25481–25489. https://doi.org/10.1074/jbc.M301403200
  51. Maslowski KM, Vieira AT, Ng A et al (2009) Regulation of inflammatory responses by gut microbiota and chemoattractant receptor GPR43. Nature 461(7268):1282–1286. https://doi.org/10.1038/nature08530
  52. Hirschberg S, Gisevius B, Duscha A, Haghikia A. (2019) Implications of diet and the gut microbiome in neuroinflammatory and neurodegenerative diseases. Int J Mol Sci 20:3109. https://doi.org/10.3390/ijms20123109
  53. Lavelle A, Sokol H (2020) Gut microbiota-derived metabolites as key actors in inflammatory bowel disease. Nat Rev Gastroenterol Hepatol 17(4):223–237. https://doi.org/10.1038/s41575-019-0258-z
  54. Duscha A, Gisevius B, Hirschberg S et al (2019) Propionic acid shapes the multiple sclerosis disease course by an immunomodulatory mechanism. Cell 180:1067.e16–1080.e16. https://doi.org/10.1016/j.cell.2020.02.035
  55. Arpaia N, Campbell C, Fan X, Dikiy S, van der Veeken J, deRoos P, Liu H, Cross JR, Pfeffer K, Coffer PJ, Rudensky AY (2013) Metabolites produced by commensal bacteria promote peripheral regulatory T-cell generation. Nature 504:451–455. https://doi.org/10.1038/nature12726
  56. Duscha A, Hegelmaier T, Dürholz K, Desel C, Gold R, Zaiss MM, Haghikia A (2022) Propionic acid beneficially modifies osteoporosis biomarkers in patients with multiple sclerosis. Ther Adv Neurol Disord 15:17562864221103935. https://doi.org/10.1177/17562864221103935
  57. Cummings JH, Pomare EW, Branch WJ, Naylor CP, Macfarlane GT (1987) Short chain fatty acids in human large intestine hepatic and venous blood. Gut 28:1221–7. https://doi.org/10.1136/gut.28.10.1221
  58. Yuille S, Reichardt N, Panda S, Dunbar H, Mulder IE (2018) Human gut bacteria as potent class I histone deacetylase inhibitors in vitro through production of butyric acid and valeric acid. PLoS One 13(7):e0201073. https://doi.org/10.1371/journal.pone.0201073
  59. Gio-Batta M, Spetz K, Barman M, Bråbäck L, Norin E, Björkstén B, Wold AE, Sandin A (2022) Low Concentration of Fecal Valeric Acid at 1 Year of Age Is Linked with Eczema and Food Allergy at 13 Years of Age: Findings from a Swedish Birth Cohort. Int Arch Allergy Immunol 183(4):398–408. https://doi.org/10.1159/000520149
  60. Wolin MJ, Miller TL, Yerry S, Zhang Y, Bank S, Weaver GA (1999) Changes of fermentation pathways of fecal microbial communities associated with a drug treatment that increases dietary starch in the human colon. Appl Environ Microbiol 65(7):2807–12. https://doi.org/10.1128/AEM.65.7.2807-2812.1999
  61. Simpson HL, Campbell BJ (2015) Review article: dietary fibre-microbiota interactions. Aliment Pharmacol Ther 42(2):158–79. https://doi.org/10.1111/apt.13248
  62. LeBlanc JG, Chain F, Martín R, Bermúdez-Humarán LG, Courau S, Langella P (2017) Beneficial effects on host energy metabolism of short-chain fatty acids and vitamins produced by commensal and probiotic bacteria. Microb Cell Fact 16(1):79. https://doi.org/10.1186/s12934-017-0691-z
  63. Meimandipour A, Hair-Bejo M, Shuhaimi M, Azhar K, Soleimani AF, Rasti B, Yazid AM (2010) Gastrointestinal tract morphological alteration by unpleasant physical treatment and modulating role of Lactobacillus in broilers. Br Poult Sci 51:52–59. https://doi.org/10.1080/00071660903394455
  64. Martin-Gallausiaux C, Marinelli L, Blottière HM, Larraufie P, Lapaque N (2021) SCFA: mechanisms and functional importance in the gut. Proc Nutr Soc 80(1):37–49. https://doi.org/10.1017/S0029665120006916
  65. He J, Xu S, Zhang B, Xiao C, Chen Z, Si F, Fu J, Lin X, Zheng G, Yu G, Chen J (2020) Gut microbiota and metabolite alterations associated with reduced bone mineral density or bone metabolic indexes in postmenopausal osteoporosis. Aging (Albany NY) 12(9):8583–8604. https://doi.org/10.18632/aging.103168
  66. Zaiss MM, Jones RM, Schett G, Pacifici R (2019) The gut-bone axis: how bacterial metabolites bridge the distance. J Clin Invest 15;129(8):3018–3028. https://doi.org/10.1172/JCI128521
  67. Roberfroid M, Gibson GR, Hoyles L et al (2010) Prebiotic effects: metabolic and health benefits. Br J Nutr 104 Suppl 2:S1-63. https://doi.org/10.1017/S0007114510003363
  68. Yan J, Charles JF (2017) Gut Microbiome and Bone: to Build, Destroy, or Both? Curr Osteoporos Rep 15(4):376–384. https://doi.org/10.1007/s11914-017-0382-z
  69. Yan J, Takakura A, Zandi-Nejad K, Charles JF (2017) Mechanisms of gut microbiota-mediated bone remodeling. Gut Microbes 9(1):84–92. https://doi.org/10.1080/19490976.2017.1371893
  70. Morrison DJ, Preston T (2016) Formation of short chain fatty acids by the gut microbiota and their impact on human metabolism. Gut Microbes 7(3):189–200. https://doi.org/10.1080/19490976.2015.1134082
  71. D'Amelio P, Sassi F (2018) Gut Microbiota, Immune System, and Bone. Calcif Tissue Int. 102(4):415–425. https://doi.org/10.1007/s00223-017-0331-y
  72. D’Amelio P, Grimaldi A, Di Bella S et al (2008) Estrogen deficiency increases osteoclastogenesis up-regulating T cells activity: a key mechanism in osteoporosis. Bone 43:92–100. https://doi.org/10.1016/j.bone.2008.02.017
  73. Ratajczak W, Rył A, Mizerski A, Walczakiewicz K, Sipak O, Laszczyńska M (2019) Immunomodulatory potential of gut microbiome-derived short-chain fatty acids (SCFA). Acta Biochim Pol 66(1):1–12. https://doi.org/10.18388/abp.2018_2648
  74. Chang PV, Hao L, Offermanns S, Medzhitov R (2014) The microbial metabolite butyrate regulates intestinal macrophage function via histone deacetylase inhibition. Proc Natl Acad Sci U S A 111(6):2247–2252. https://doi.org/10.1073/pnas.1322269111
  75. Huang J, Wang L, Dahiya S, Beier UH, Han R, Samanta A, Bergman J, Sotomayor EM, Seto E, Kozikowski AP, Hancock WW (2017) Histone/protein deacetylase 11 targeting promotes Foxp3+ Treg function. Sci Rep 7(1):8626. https://doi.org/10.1038/s41598-017-09211-3
  76. Montalvany-Antonucci CC, Duffles LF, de Arruda JAA, Zicker MC, de Oliveira S, Macari S, Garlet GP, Madeira MFM, Fukada SY, Andrade I Jr, Teixeira MM, Mackay C, Vieira AT, Vinolo MA, Silva TA (2019) Short-chain fatty acids and FFAR2 as suppressors of bone resorption. Bone 125:112–121. https://doi.org/10.1016/j.bone.2019.05.016
  77. Park J, Kim M, Kang SG, Jannasch AH, Cooper B, Patterson J, Kim CH (2015) Short-chain fatty acids induce both effector and Tregs by suppression of histone deacetylases and regulation of the mTOR-S6K pathway. Mucosal Immunol 8:80–93. https://doi.org/10.1038/mi.2014.44
  78. Trompette A, Gollwitzer ES, Yadava K et al (2014) Gut microbiota metabolism of dietary fiber influences allergic airway disease and hematopoiesis. Nat Med 20:159–166. https://doi.org/10.1038/nm.3444
  79. Zhao SC, Teng XQ, Xu DL, Chi X, Ge M, Xu SW (2020) Influences of low level of dietary calcium on bone characters in laying hens. Poult Sci 99(12):7084–7091. https://doi.org/10.1016/j.psj.2020.08.057
  80. Rovenský J, Stancíková M, Masaryk P, Svík K, Istok R (2003) Eggshell calcium in the prevention and treatment of osteoporosis. Int J Clin Pharmacol Res 23(2–3):83–92. https://doi.org/10.1109/TNSRE.2008.2010475
  81. Quigley EMM (2019) Prebiotics and Probiotics in Digestive Health. Clin Gastroenterol Hepatol. 17(2):333–344. https://doi.org/10.1016/j.cgh.2018.09.028
  82. Weaver CM (2015) Diet, gut microbiome, and bone health. Curr Osteoporos Rep 13(2):125–30. https://doi.org/10.1007/s11914-015-0257-0
  83. Whisner CM, Martin BR, Nakatsu CH, McCabe GP, McCabe LD, Peacock M, Weaver CM (2014) Soluble maize fibre affects short-term calcium absorption in adolescent boys and girls: a randomised controlled trial using dual stable isotopic tracers. Br J Nutr 112(3):446–56. https://doi.org/10.1017/S0007114514000981
  84. Wallace TC, Marzorati M, Spence L, Weaver CM, Williamson PS (2017) New frontiers in fibers: innovative and emerging study on the gut microbiome and bone health. J Am Coll Nutr 36:218–222. https://doi.org/10.1080/07315724.2016.1257961
  85. Wallimann A, Magrath W, Pugliese B et al (2021) Butyrate Inhibits Osteoclast Activity In Vitro and Regulates Systemic Inflammation and Bone Healing in a Murine Osteotomy Model Compared to Antibiotic-Treated Mice. Mediators Inflamm 2021:8817421. https://doi.org/10.1155/2021/8817421
  86. Gultemirian ML, Corti HR, Chaia AP et al (2014) Fermentation in vitro of a mixture of dietary fibers and cane molasses by the cecal microbiota: Application on mineral absorption through the laying hen's colonic epithelium. Animal Feed Science & Technology 191:76–82. https://doi.org/10.1016/j.anifeedsci.2014.01.019
  87. Tyagi AM, Yu M, Darby TM, Vaccaro C, Li JY, Owens JA, Hsu E, Adams J, Weitzmann MN, Jones RM, Pacifici R (2018) The microbial metabolite butyrate stimulates bone formation via T regulatory cell-mediated regulation of WNT10B expression. Immunity 49(6):1116–1131. https://doi.org/10.1016/j.immuni.2018.10.013
  88. Wang Y, Li N, Yang JJ, Zhao DM, Chen B, Zhang GQ, Chen S, Cao RF, Yu H, Zhao CY, Zhao L, Ge YS, Liu Y, Zhang LH, Hu W, Zhang L, Gai ZT (2020) Probiotics and fructo-oligosaccharide intervention modulate the microbiota-gut brain axis to improve autism spectrum reducing also the hyper-serotonergic state and the dopamine metabolism disorder. Pharmacol Res 157:104784. https://doi.org/10.1016/j.phrs.2020.104784
  89. De Vernejoul MC, Collet C, Chabbi-Achengli Y (2012) Serotonin: good or bad for bone. Bonekey Rep 1:120. https://doi.org/10.1038/bonekey.2012.120
  90. Ducy P (2011) 5-HT and bone biology. Curr Opin Pharmacol 11(1):34–8. https://doi.org/10.1016/j.coph.2011.01.007
  91. Reigstad CS, Salmonson CE, Rainey JF 3rd, Szurszewski JH, Linden DR, Sonnenburg JL, Farrugia G, Kashyap PC (2015) Gut microbes promote colonic serotonin production through an effect of short-chain fatty acids on enterochromaffin cells. FASEB J 29:1395–1403. https://doi.org/10.1096/fj.14-259598
  92. Katono T, Kawato T, Tanabe N, Suzuki N, Iida T, Morozumi A, Ochiai K, Maeno M (2008) Sodium butyrate stimulates mineralized nodule formation and osteoprotegerin expression by human osteoblasts. Arch Oral Biol 53(10):903–9. https://doi.org/10.1016/j.archoralbio.2008.02.016
  93. Ko B (2017) Parathyroid hormone and the regulation of renal tubular calcium transport. Curr Opin Nephrol Hypertens 26(5):405–410. https://doi.org/10.1097/MNH.0000000000000347
  94. Smith PM, Howitt MR, Panikov N, Michaud M, Gallini CA, Bohlooly-Y M, Glickman JN, Garrett WS (2013) The microbial metabolites, short-chain fatty acids, regulate colonic Treg cell homeostasis. Science 341(6145):569–573. https://doi.org/10.1126/science.1241165
  95. Liu F, Kong A, Fu P, Cao QQ, Tao KS, Liu DY, Wang XB, Tong ZX, Rehman MU, Huang SC (2021) Lactobacillus rhamnosus JYLR-005 Prevents Thiram-Induced Tibial Dyschondroplasia by Enhancing Bone-Related Growth Performance in Chickens. Probiotics Antimicrob Proteins 13(1):19–31. https://doi.org/10.1007/s12602-020-09670-7
  96. Tyagi AM, Yu M, Darby TM, Vaccaro C, Li JY, Owens JA, Hsu E, Adams J, Weitzmann MN, Jones RM, Pacifici R (2018) The Microbial Metabolite Butyrate Stimulates Bone Formation via T Regulatory Cell-Mediated Regulation of WNT10B Expression. Immunity 18;49(6):1116–1131.e7. https://doi.org/10.1016/j.immuni.2018.10.013
  97. Doublier S, Cirrincione S, Scardaci R, Botta C, Lamberti C, Giuseppe FD, Angelucci S, Rantsiou K, Cocolin L, Pessione E (2022) Putative probiotics decrease cell viability and enhance chemotherapy effectiveness in human cancer cells: role of butyrate and secreted proteins. Microbiol Res 260:127012. https://doi.org/10.1016/j.micres.2022.127012
  98. Montalvany-Antonucci CC, Duffles LF, de Arruda JAA, Zicker MC, de Oliveira S, Macari S, Garlet GP, Madeira MFM, Fukada SY, Andrade I Jr, Teixeira MM, Mackay C, Vieira AT, Vinolo MA, Silva TA (2019) Short-chain fatty acids and FFAR2 as suppressors of bone resorption. Bone 125:112–121. https://doi.org/10.1016/j.bone.2019.05.016
  99. Lemma S, Sboarina M, Porporato PE, Zini N, Sonveaux P, Di Pompo G, Baldini N, Avnet S (2016) Energy metabolism in osteoclast formation and activity. Int J Biochem Cell Biol 79:168–180. https://doi.org/10.1016/j.biocel.2016.08.034
  100. Lucas S, Omata Y, Hofmann J, Böttcher M, Iljazovic A, Sarter K, Albrecht O, Schulz O, Krishnacoumar B, Krönke G, Herrmann M, Mougiakakos D, Strowig T, Schett G, Zaiss MM (2018) Short-chain fatty acids regulate systemic bone mass and protect from pathological bone loss. Nat Commun 4;9(1):55. https://doi.org/10.1038/s41467-017-02490-4
  101. Wauquier F, Philippe C, Léotoing L, Mercier S, Davicco MJ, Lebecque P, Guicheux J, Pilet P, Miot-Noirault E, Poitout V, Alquier T, Coxam V, Wittrant Y (2013) The free fatty acid receptor G protein-coupled receptor 40 (GPR40) protects from bone loss through inhibition of osteoclast differentiation. J Biol Chem 288(9):6542–51. https://doi.org/10.1074/jbc.M112.429084
  102. Scott KP, Martin JC, Campbell G, Mayer CD, Flint HJ (2006) Whole-genome transcription profiling reveals genes up-regulated by growth on fucose in the human gut bacterium "Roseburia inulinivorans". Journal of Bacteriology 188(12):4340–9. https://doi.org/10.1128/JB.00137-06
  103. Rey F E, Faith JJ, Bain J, Muehlbauer MJ, Stevens RD, Newgard CB, Gordon JI (2010) Dissecting the in Vivo Metabolic Potential of Two Human Gut Acetogens. Journal of Biological Chemistry 285(29):22082–90. https://doi.org/10.1074/jbc.M110.117713
  104. Van den Abbeele P, Belzer C, Goossens M, Kleerebezem M, De Vos WM, Thas O, De Weirdt R, Kerckhof FM, Van de Wiele T (2013) Butyrate-producing Clostridium cluster XIVa species specifically colonize mucins in an in vitro gut model. Isme Journal 7(5):949–61. https://doi.org/10.1038/ismej.2012.158
  105. Shetty SA, Marathe NP, Lanjekar V, Ranade D, Shouche YS (2013) Comparative Genome Analysis of Megasphaera sp Reveals Niche Specialization and Its Potential Role in the Human Gut. Plos One 8(11). https://doi.org/10.1371/journal.pone.0079353
  106. Reichardt N, Duncan SH, Young P, Belenguer A, McWilliam Leitch C, Scott KP, Flint HJ, Louis P (2014). Phylogenetic distribution of three pathways for propionate production within the human gut microbiota. ISME J 8(6):1323–35. https://doi.org/10.1038/ismej.2014.14
  107. Tang WHW, Li DY, Hazen SL (2019) Dietary metabolism, the gut microbiome, and heart failure. Nat Rev Cardiol 16(3):137–154. https://doi.org/10.1038/s41569-018-0108-7
  108. Hayashi H, Shibata K, Sakamoto M, Tomita S, Benno Y (2007) Prevotella copri sp. nov. and Prevotella stercorea sp. nov., isolated from human faeces. Int J Syst Evol Microbiol 57(Pt 5):941–6. https://doi.org/10.1099/ijs.0.64778-0
  109. Louis P, Hold GL, Flint HJ (2014) The gut microbiota, bacterial metabolites and colorectal cancer. Nat Rev Microbiol 12(10):661–72. https://doi.org/10.1038/nrmicro3344
  110. Zhang WH, Jiang Y, Zhu QF, Gao F, Dai S F, Chen J, Zhou G H (2011) Sodium butyrate maintains growth performance by regulating the immune response in broiler chickens. British poultry science 52(3):292–301. https://doi.org/10.1080/00071668.2011.578121
  111. Zou X, Ji J, Qu H, Wang J, Shu DM, Wang Y, Liu TF, Li Y, Luo CL (2019) Effects of sodium butyrate on intestinal health and gut microbiota composition during intestinal inflammation progression in broilers. Poultry science 98(10):4449–4456. https://doi.org/10.3382/ps/pez279
  112. Zhao X, Wang Y, Nie Z, Han L, Zhong X, Yan X, Gao X (2020) Eucommia ulmoides leaf extract alters gut microbiota composition, enhances short-chain fatty acids production, and ameliorates osteoporosis in the senescence-accelerated mouse P6 (SAMP6) model. Food Sci Nutr 8(9):4897–4906. https://doi.org/10.1002/fsn3.1779
  113. Wallimann A, Magrath W, Pugliese B, Stocker N, Westermann P, Heider A, Gehweiler D, Zeiter S, Claesson M J, Richards RG, Akdis CA, Hernandez CJ, O'Mahony L, Thompson K, Moriarty TF (2021) Butyrate Inhibits Osteoclast Activity In Vitro and Regulates Systemic Inflammation and Bone Healing in a Murine Osteotomy Model Compared to Antibiotic-Treated Mice. Mediators Inflamm 2021:8817421. https://doi.org/10.1155/2021/8817421
  114. Weaver CM, Martin BR, Story JA, Hutchinson I, Sanders L (2010) Novel fibers increase bone calcium content and strength beyond efficiency of large intestine fermentation. J Agric Food Chem 58(16):8952–8957. https://doi.org/10.1021/jf904086d
  115. Rahman MM, Kukita A, Kukita T, Shobuike T, Nakamura T, Kohashi O (2003) Two histone deacetylase inhibitors, trichostatin A and sodium butyrate, suppress differentiation into osteoclasts but not into macrophages. Blood 101(9):3451–3459. https://doi.org/10.1182/blood-2002-08-2622
  116. Cho KM, Kim YS, Lee M, Lee HY, Bae YS (2021) Isovaleric acid ameliorates ovariectomy-induced osteoporosis by inhibiting osteoclast differentiation. J Cell Mol Med 25(9):4287–4297. https://doi.org/10.1111/jcmm.16482

Tables

Table 1Gut microbiota for the synthesis of short-chain fatty acids.

Phylum

Class

Order

Family

Genus/Species

SCFAs

Reference

Actinobacteria

Actinobacteria

Bifidobacteriales

Bifidobacteriaceae

Bifidobacterium adolescentis

Propionic acid

[106]

 

 

 

 

Bifidobacterium sp.

Acetic acid

Bacteroidetes

Bacteroidetes

Bacteroidetes

Bacteroidaceae

Bacteroides fragilis

Propionic acid Butyric acid

[102, 106, 107]

 

 

 

 

Bacteroides sp.

Acetic acid Propionic acid

 

 

 

 

Bacteroides thetaiotaomicron

Propionic acid Butyric acid

 

Bacteroidetes

Bacteroidetes

Prevotellaceae

Prevotella stercorea

Acetic acid

Valeric acid

[108]

Firmicutes

Bacilli

Lactobacillales

Streptococcaceae

Streptococcus sp.

Acetic acid

[102, 109]

 

Clostridia

Clostridiales

Clostridiaceae

Clostridium beijerickii

Propionic acid Butyric acid

[102, 103, 104, 106, 107, 109]

 

 

 

 

Clostridium botulinum

Propionic acid Butyric acid

 

 

 

 

Clostridium butyricum

Butyric acid

 

 

 

Lachnospiraceae

Coprococcus comes

Propionic acid Butyric acid

 

 

 

 

 

Coprococcus eutactus

Propionic acid Butyric acid

 

 

Vellionellales

Veillonellaceae

Megasphaera elsdenii

Acetic acid Propionic acid Butyric acid Valeric acid

[105, 109]

 

 

 

 

Megasphaera sp.

Acetic acid Propionic acid Butyric acid Valeric acid

Proteobacteria

Gammaproteo

bacteria

Enterobacteriales

Enterobacteriaceae

Salmonella sp.

Propionic acid

[109]

Verrucomicrobia

Verrucomicrobia

Verrucomicrobiales

Verrucomicrobiaceae

Akkermansia muciniphila

Acetic acid Propionic acid

[106]

Note: Only part of the gut microbiota that can produce SCFA is listed in the table

Table 2. The function and role of SCFA.

Function

Types of SCFA

Object of study

Effect

Reference

Immunity

Butyric acid

Broilers

The addition of sodium butyrate to broiler diets can improve the growth performance of broilers and can be used to regulate the body's immune response and help to reduce the production of bone inflammation.

[110]

Butyric acid

Broilers

Butyric acid as a feed additive can significantly reduce the level of IL-6 and IL-1β, relieve the development and spread of inflammation, and facilitate the growth and development of broilers.

[111]

Acetic acid

Butyric acid

Isobutyric acid

Mice

The production of SCFAs has a mitigating effect on OP development.

[112]

The regulation of calcium

Acetic acid

Propionic acid

Butyric acid

Laying hens

Promote intestinal absorption of calcium.

[86]

Acetic acid

Propionic acid

Butyric acid

Valeric acid

Mice

Increases calcium deposition at the site of bone injury and accelerates bone formation

[113]

Acetic acid

Propionic acid 

Butyric acid

Isobutyric acid

Valeric acid 

Isovaleric acid

Rats

Increases mineral availability by increasing calcium dissolution at lower pH, thereby increasing bone mineral content and deposition.

[114]

Osteogenesis

Butyric acid

Human beings

Increased mineralization of osteoblasts promotes bone formation and inhibits osteoclast differentiation by promoting OPG production by human osteoblasts

[93]

Butyric acid

Mice

Butyrates can increase the number of Tregs in the intestine and bone marrow. Tregs can stimulate CD8 + T cells, which can secrete Wnt10b and promote bone formation by activating Wnt signalling in osteoblasts.

[55,87]

Propionic acid

SPF Mice

Propionate can promote the proliferation of Tregs, thereby enhancing the regulation of Tregs on bone homeostasis.

[95]

Butyric acid

Mice

Induced proliferation of Tregs in the gut and bone tissue and increased trabecular bone volume, and promoted bone formation in mice.

[96]

Osteoclastogenesis

Butyric acid

Rats, Mice

Butyrate can inhibit the production of osteoclast precursor cells by inhibiting the activity of Histone deacetylase (HDAC). Butyric acid inhibits the formation of osteoclasts and the expression of osteoclast-specific mRNA under the stimulation of RANKL.

[115]

Isovaleric acid

Mice

Isovaleric acid suppresses differentiation of bone marrow-derived macrophages into OCs by RANKL. Isovaleric acid inhibited the expression of OC-related genes.

[116]

Acetic acid

Propionic acid

Butyric acid

Valeric acid

Mice

Genes related to osteoclast differentiation are differentially expressed in osteoclast precursor cells, which can significantly reduce osteoclast formation and bone resorption activity.

[85]

Note: OP, Osteoporosis; HDAC, Histone Deacetylase; RANKL, Receptor Activator of Nuclear Factor-κB Ligand.