Skip to main content

The ameliorative role of specific probiotic combinations on bone loss in the ovariectomized rat model

Abstract

Background

Osteoporosis, a skeletal disease described by impaired bone strength, cause an increased risk of fractures. We aimed in this study to clarify which particular wise combination of probiotics has the most beneficial effect in the rat model of osteoporosis.

Methods

Sixty-three mature female Sprague Dawley rats (12–14 weeks old, weight 200 ± 20 g) were ovariectomized and then divided into nine random groups, each group consisting of 7 rats. Lactic acid bacteria were isolated from traditional fermented yogurt on the northern coast of the Persian Gulf. Seven combinations of probiotics, each containing three probiotic strains, were designed and administered (1 × 10 9 CFU / ml/strain daily along with their water) to treat ovariectomized rats. The period from ovariectomy to eutanásia was 3 months. For evaluating femur, spine, and tibia, bone mineral density (BMD), and bone mineral content (BMC), Dual-energy X-ray absorptiometry (DEXA) scans were performed. Also, effect of probiotic combinations was assessed on biochemical markers including vitamin D, calcium, phosphorus, and alkaline phosphatase in serum.

Results

Combination NO 4, containing L. acidophilus, B. longum, and L. reuteri, is the most influential group on global, spine, and femur BMD. Combination NO 3, containing L. acidophilus, L. casei, and L. reuteri, also significantly affects the BMD of the tibia among the treatment group. We found that the combination NO 4 had the most significant ameliorative effect on global BMC. Also, combination NO 1 (comprising L. acidophilus, L. casei, and B. longum), NO 6 (containing L. casei, B. longum, and Bacillus coagulans), NO 7 (containing L. casei, L. reuteri, and B. longum), and NO 4 had the most considerable raising effect on spine BMC. In addition, the serum calcium and Vitamin D concentration in the groups NO 4, 6, and 7 were significantly higher than in OVX groups, whereas the alkaline phosphatase concentration was considerably reduced in these groups.

Conclusion

Among nine effective probiotics, a combination containing L. acidophilus, B. longum, and L. reuteri is the most influential group in ovariectomized osteoporotic rat.

Peer Review reports

Introduction

Osteoporosis, a skeletal disease described by impaired bone strength that causes an increased risk of fractures, is a worldwide disease that mainly occurs in postmenopausal women and older adults [1, 2]. The condition is a bothering factor for about 200 million people, with low bone mass, worsening of the fine structure of bone tissue, and elevated chance of fractures [3]. Since the population is getting old, it is predictable that the prevalence of osteoporosis will significantly increase [4].

Probiotics are living microorganisms that bring health benefits to consumers by promoting intestinal balance [5, 6]. They contain different microbes explained by their genus, species, and strain designation [7]. Probiotic supplements are beneficial for humans and animals and can be used after antibiotic therapy to compensate for gut microbiome deficiency or as prophylaxis [8]. The International Scientific Association of Probiotics and Prebiotics (ISAPP) has a helpful definition of probiotics consisting of three properties; probiotics should be alive during administration, be healthy for users, and have the right dose when delivered [9].

Many experiments have been recently done to specify the potential profits of probiotic preparations in pathologic bone disorders such as osteoporosis. In this regard, several probiotic strains have been introduced in a single administration or as food supplementation to ameliorate bone loss [2]. Oral intake of a single probiotic strain Limosilactobacillus reuteri (L. reuteri) for 28 days by male rat caused substantial health benefits for trabecular bone density, number, thickness, and bone mineral content and density both in the femur and vertebral [10]. A study showed that when a single strain of Bifidobacterium longum (B. Longum) was orally administered to male rats for 4 weeks, the microelements essential for bone health, such as calcium, phosphorus, and magnesium, were concentrated in the tibia [11]. Also, Lacticaseibacillus paracasei (L. paracasei) could increase cortical bone mineral content, and the resorption marker C-terminal telopeptides in rat blood were decreased as well as calcium exertion through urine [12]. This probiotic strain stimulates differentiation of osteoblasts via bone morphogenetic protein (BMP) and inhibits RANKL-induced differentiation of osteoclast, thus helping to inhibit bone loss. Dar et al. reported that Lactobacillus acidophilus (L. acidophilus) improves the microarchitecture of both trabecular bone and cortical bone, enhancing bone mineral density and heterogeneity immunomodulatory impact on the host immune system [13]. Also, we observed that the supernatant of Bacillus coagulans (B. coagulans) enriched the tibia bone mineral density (BMD) of OVX rat in the previous study [2]. In addition, our previous studies evaluated the effects of isolated several native strains of probiotics on bone loss in ovariectomized rats [3]. Although their mechanism of action hasn’t been entirely understood, these probiotics can affect the regulation of luminal pH, production of enzymes, organic acids, and antimicrobial peptides, improvement of barrier function by enriching mucus secretion, stimulation of osteoblast differentiation, and maintaining of the host immune system [2, 14]. They also may have effects on preventing and treating osteoporosis by affecting calcium absorption via decreasing pH, inhibiting calcium binding to bile acid and elevating the surface area for absorption in the large intestine, regulating the immune responses, and producing small molecules, for instance, serotonin or estrogen-like molecules [15,16,17].

Combining several probiotic strains appears to have greater efficacy against many diseases than single-strain therapy. Few studies have shown that multi-strain probiotics, including the strains that are a component of the combination itself, are more potent [18]. However, it is still unclear whether this superiority is because of synergistic effects between the strains or because of the high dose of probiotics used.

To accurately demonstrate the greater efficacy of multivariate probiotics and which multivariate probiotic compound is more effective for treating osteoporosis, further studies using the same doses in similar populations are needed. Given the positive effect of probiotics alone or in randomized combination in the treatment of osteoporosis, we aimed in this study to clarify which particular wise combination of probiotics has the most beneficial effect in the postmenopausal rat model. Therefore, due to the positive impact of probiotics alone in the treatment of osteoporosis, we configured them in seven selective combinations and compared their effectiveness in protecting rats from ovariectomized (OVX)-induced bone loss. As there are several probiotic combinations in the supplements market, this study aimed to determine which probiotic combination is more effective in ameliorating bone loss in OVX rats using quantitative indicators.

Material and methods

Isolation and formulation of bacteria

Twenty samples of traditional fermented yogurt from the northern coast of the Persian Gulf were used for lactic acid bacterial isolation. After preparation, the samples were stored in a refrigerator at 4 °C. 10 g of each was diluted in peptone water (4%), homogenized with a laboratory mixer, and serially diluted with sterile water. To count the Lactobacillus, Streptococcus, and Bifidobacterium, LS differential medium was used. De Man, Rogosa, Sharpe (MRS) agar, and Bifidobacterium medium (BFM) agar isolate Lactobacilli and Bifidobacteria. All plates were incubated under anaerobic conditions at 37° C for three days. According to Bergey's Manual of Systematic Bacteriology, MRS and BFM agar isolates were identified based on cultural, morphological, and biochemical properties. After identification, the strain was maintained by subculture in tryptic soybean agar (TSA) medium at 37° C and stored in tryptic soybean broth (TSB) medium at 4° C until lyophilization (maximum storage days was two weeks). Before use, the lyophilized strain was formulated with phosphate-buffered saline (PBS, pH 7.4) and mechanically stirred for 15 min to mix well. The probiotic PBS solution premix was prepared for oral tube feeding. The concentration of probiotic candidates at each interval was 1.5 × 108 colony forming units (CFU) / ml.

An equal amount of each strain mentioned in Table 1 was used to prepare the probiotic combinations to reach the final concentration of 109. To ensure that our combination samples were qualified and had no contaminant, every sample given to the animals was cultured.

Table 1 Content of specific combinations of probiotics used in this study

Experimental design

Sixty-three adult female Sprague Dawley rats (12–14 weeks old, weight 200 ± 20 g) were purchased from the Laboratory Animal Center of Shiraz Medical University. Rats were brought up under typical laboratory conditions (room temperature (23 ± 2° C), 60 ± 5% relative humidity, and 12/12-h light / dark cycle) with a standard pellet diet and water and given freely. A standard pellet diet composition was as follows: Crude protein 23%, crude fat 3.5%, crude fiber 4.5%, ash 10%, calcium 0.95–1%, phosphorus 0.65–0.7%, NaCl 0.5%, lysine 1.15%, methionine 0.33%, threonine 0.72%, tryptophan 0.25%, cysteine 0.3%). Rats were acclimatized to the animal room for one week. They were then divided into nine random groups, each consisting of 7 rats. The classification of animals is according to this:

  • group 1, control;

  • group 2, OVX;

  • group 3, OVX + combination NO 1;

  • group 4, OVX + combination NO 2;

  • group 5, OVX + combination NO 3;

  • group 6, OVX + combination NO 4;

  • group 7, OVX + combination NO 5,

  • group 8, OVX + combination NO 6,

  • group 9, OVX + combination NO 7.

The contents of each combination are mentioned in Table 1.

The animals in groups 3, 4, 5, 6, 7, 8, and 9 were treated with 1 ml (1 × 10 9 CFU / ml/strain daily along with their water) of probiotics for four weeks. Normal saline was supplied to the rats of groups 1 and 2. Food and water absorption were monitored and did not differ between groups. This work was approved by the Ethics Committee (NO. IR.SUMS.REC.1398.500) of Shiraz University of Medical Sciences, Shiraz, Iran.

Ovarectomy procedure

Adult female rats were bilaterally ovariectomized. The period of ovariectomy until euthanasia was 3 months. Ketamine 10% (100 mg/kg, Alfasan, Netherlands) and xylazine 2% (10 mg/kg, Alfasan, Netherlands) were the anesthetics used in this study. Both ovaries were resected in all groups, except for the first group, which was the control group, after surgical anastomosis of the uterine horns through a central longitudinal incision. Sham operation was performed in the control group.

Dual-energy X-ray absorptiometry parameter measurements

In order to assess the area, bone mineral content (BMC), and bone mineral density (BMD) of femur, spine, and tibia, Dual-energy X-ray absorptiometry (DXA) scans were applied on a Discovery QDR, USA device with Hologic instrument via the particular software for small animals at the experiment termination. At first, we set up the RAT STEP PHANTOM (Hologic P/N010-0758Rev.004) scan. In this method, when the system motion was completed, we centered the STEP PHANTOM on the table along the long axis of the laser with the cross-hair ¾ "(2 cm) of the right edge of the thinnest step. Then we pressed a continue button to start the scan. BMC in grams, bone area (BA) in square centimeters, and BMD in g/cm2 were measured.

Biochemical analysis of serum

Blood samples were collected in chilled non-heparinized tubes for clotting at room temperature by cardiocentesis. Then they were centrifuged at 3500 rpm at 4 ͦC for 20 min, and biochemical markers including vitamin D, calcium (Ca), phosphorus (P), and alkaline phosphatase (ALP) were measured by assessing isolated sera (Fig. 3).

Statistical analysis

Statistical analysis was performed using IBM © SPSS © Statistics v 22.0 for Windows. The data are expressed as mean ± standard deviation. One-way analysis of variance (ANOVA) was applied to analyze the association between existing probiotic combinations and bone mineral density measurement parameters. Tukey post hoc analysis was performed when the ANOVA outcomes exhibited significance (p ≤ 0.05).

Results

Bacterial strains isolation and identification

The physiological and biochemical properties of the five bacterial strains applied in this study are mentioned individually in Table 2. The isolated probiotics are Lactobacillus acidophilus, Limosilactobacillus reuteri, Lacticaseibacillus casei, Bifidobacterium longum, and Bacillus coagulans (previously known as Lactobacillus sporogenes) based on the standard references and morphological characteristics.

Table 2 The physiological and biochemical properties of the selected strains

Effect of probiotic combinations on bone mineral density

Figure 1 displays the effect of the probiotic combination on the overall spine, femur, and tibia BMD. Combinations # 4, 6, and 7 significantly ameliorated the global BMD of the OVX-treated group compared to the untreated OVX group. Combinations NO 1, 2, 3, and 5 had no significant increasing effect on the global BMD (Fig. 1a). Spine BMD of treated OVX groups was significantly increased when treated with all of the probiotic combinations vs. untreated OVX group (Fig. 1b). Combinations NO 2, 3, 4, and 7 increased the femur BMD remarkably in the treated OVX groups in comparison with the untreated group. Combination NO 1, 5, and 6 had no considerable effect on treated groups (Fig. 1c). In terms of tibia BMD, combinations NO 3 and 4 considerably increased BMD vs. untreated OVX group. In contrast, combinations NO 6 and 7 had a minor effect on BMD (Fig. 1d).

Fig. 1
figure 1

The effect of the probiotic combinations on the overall spine, femur, and tibia BMD. Treat 1: OVX + combination of Lactobacillus acidophilus, Lacticaseibacillus casei, Bifidobacterium longum; Treat 2: OVX + combination of Lactobacillus acidophilus, Lacticaseibacillus casei, Bacillus coagulans, Treat 3: OVX + combination of Lactobacillus acidophilus, Lacticaseibacillus casei, Limosilactobacillus reuteri; Treat 4: OVX + combination of Lactobacillus acidophilus, Bifidobacterium longum, Limosilactobacillus reuteri; Treat 5: OVX + combination of Lactobacillus acidophilus, Bifidobacterium longum, Bacillus coagulans; Treat 6: OVX + combination of Lacticaseibacillus casei, Bifidobacterium longum,Bacillus coagulans; Treat 7: OVX + combination of Lacticaseibacillus casei, Limosilactobacillus reuteri, Bifidobacterium longum. There were no significant differences between columns, which have at least one similarly letters. However, dissimilar letters indicate significance (P < 0.05)

Effect of probiotic combinations on bone mineral content (BMC)

Figure 2 shows the effect of a probiotic combination on global, spine, femur, and tibia bone mineral content (BMC). As exists in Fig. 2a, all combinations significantly augmented the global BMC of OVX groups compared to untreated. Spine BMC in groups treated with combinations NO 1, 4, and 6 have significant differences from the untreated group (Fig. 2b). In the femur, the combinations NO 3, 4, 6, and 7 significantly increased the BMC (Fig. 2c). The BMC of the tibia was considerably increased while treated with combinations NO 1 to 4 and had no substantial change when treated with combinations NO 6 and 7 (Fig. 2d).

Fig. 2
figure 2

The effect of a probiotic combinations on global, spine, femur, and tibia BMC. Treat 1: OVX + combination of Lactobacillus acidophilus, Lacticaseibacillus casei, Bifidobacterium longum; Treat 2: OVX + combination of Lactobacillus acidophilus, Lacticaseibacillus casei, Bacillus coagulans, Treat 3: OVX + combination of Lactobacillus acidophilus, Lacticaseibacillus casei, Limosilactobacillus reuteri; Treat 4: OVX + combination of Lactobacillus acidophilus, Bifidobacterium longum, Limosilactobacillus reuteri; Treat 5: OVX + combination of Lactobacillus acidophilus, Bifidobacterium longum, Bacillus coagulans; Treat 6: OVX + combination of Lacticaseibacillus casei, Bifidobacterium longum,Bacillus coagulans; Treat 7: OVX + combination of Lacticaseibacillus casei, Limosilactobacillus reuteri, Bifidobacterium longum. There were no significant differences between columns, which have at least one similarly letters. However, dissimilar letters indicate significance (P < 0.05)

Effect of probiotic combinations on biochemical factors in serum

As shown in Fig. 3A, serum Ca concentration was significantly lower in the OVX group compared to the control group at the end of the experiment (P ≤ 0.006). In treated groups no. 4, 6, and 7, the Ca concentration was significantly higher compared to OVX group (Fig. 3A). These differences between group 4,6,7 and the control group were insignificant (P > 0.05). The concentration of Ca was significantly (P ≤ 0.05) lower in groups no. 1,2,3 and 5 compared to the control group, although they significantly increased Ca level compared to the OVX group (Fig. 3A).

Fig. 3
figure 3

The effect of probiotics on serum calcium (A), 1, 25 (OH)2 vitamin D (B), phosphorus (C), and alkaline phosphatase (D) concentrations in ovariectomized rats. Treat 1: OVX + combination of Lactobacillus acidophilus, Lacticaseibacillus casei, Bifidobacterium longum; Treat 2: OVX + combination of Lactobacillus acidophilus, Lacticaseibacillus casei, Bacillus coagulans, Treat 3: OVX + combination of Lactobacillus acidophilus, Lacticaseibacillus casei, Limosilactobacillus reuteri; Treat 4: OVX + combination of Lactobacillus acidophilus, Bifidobacterium longum, Limosilactobacillus reuteri; Treat 5: OVX + combination of Lactobacillus acidophilus, Bifidobacterium longum, Bacillus coagulans; Treat 6: OVX + combination of Lacticaseibacillus casei, Bifidobacterium longum,Bacillus coagulans; Treat 7: OVX + combination of Lacticaseibacillus casei, Limosilactobacillus reuteri, Bifidobacterium longum. There were no significant differences between columns, which have at least one similarly letters. However, dissimilar letters indicate significance (P < 0.05)

As expected, the serum VitD concentration was lower in the OVX group compared to the control group. Supplementation with probiotics increased VitD concentration in all combination groups, but this difference was only significant (P ≤ 0.05) in the combination groups 4,6 and 7 compared to both control and OVX groups (Fig. 3B).

The phosphorus concentration (P) was not significantly different between all experimental groups (Fig. 3C).

As the results showed, ALP concentration was significantly (P ≤ 0.05) more remarkable in the OVX group than in the control group. Treatment with combination groups 4, 6, and 7 considerably reduced ALP concentration. In other groups, ALP concentrations were elevated after treatment with probiotic combinations compared to the control group (Fig. 3D).

Discussion

Bone is a pivotal system of the human body, and its hemostasis is related to intestinal flora because it is reported that the gut microbiome can considerably affect bone physiology [19, 20]. Maintaining the gut flora equilibrium is achieved by dietary changes or using probiotics and their metabolites (oligosaccharides, carbohydrates, fibers). Probiotics can change the gut microbiota composition, induce anti-inflammatory responses, endorse intestinal calcium absorption, and thus increase BMD [20].

Previous research has unequivocally shown that the intestinal system significantly impacts bone health. One way this happens is by controlling the absorption of minerals like calcium and phosphorus, which are essential for strong bones. Additionally, incretins and serotonin, which are obtained from the gut, and endocrine variables that affect the absorption of these minerals, might affect bone turnover [21, 22]. More recent research has shown how the intestinal microbiome affects bone function using germ-free rat and probiotics [23]. Rats treated with yogurt containing L. casei, L. reuteri, and L. gasseri enhanced calcium absorption, resulting in raised BMC relative to the control in research similar to the mouse model [24]. Similarly, supplementing developing rats with L. rhamnosus (HN001) enhanced calcium retention [25]. B. longum has been found to affect bone in addition to several Lactobacillus strains positively. Male rats supplemented with B. longum (ATCC 15,707) for 28 days had a higher percentage of fracture strength and more calcium and vitamin D in their tibias than untreated rats did [11]. In a different study, rats with a high-cholesterol diet plus fermented broccoli for 12 weeks showed a decrease in the number of TRAP-positive osteoclasts compared to untreated rats [26]. The probiotic combinations used in this study (groups 4, 6, and 7) that included L. acidophilus, B. longum, L. reuteri, L. casei, and B. coagulans significantly enhanced Vitamin D and Ca absorption.

The use of probiotic compounds is believed to have several potential advantages over single-strain formulations, including a greater chance of successful treatment by increasing probiotic strains, more potential niche, and a more comprehensive range of efficacy due to the greater diversity of strains, additive or synergistic effects due to increased adhesion, creation of a favorable environment and reduce intestinal microbiota antagonism [27]. However, the probiotic combination is not always successful and can sometimes have a potentially harmful effect due to the antagonistic effects between the probiotics in the product. Therefore, designing an experiment using multidimensional settings of multi-strain probiotics for such studies can be very useful [27]. Some preliminary studies reported that a VSL#3 (containing three species of Bifidobacterium (B. longum, B. breve, and B. infantis), four strains of Lactobacillus species (L. casei, L. plantarum, L. acidophilus, and L. delbruekii subsp. bulgaricus) has been used to treat osteoporosis [28, 29]. These in-vivo studies showed that this branded combination obviously improved the femoral bone density, trabecular thickness, and number [30]. This may be due to the spinal bone volume after ovariectomy in rats treated with VSL#3.

Although numerous investigations have studied the effect of a probiotic combination in the prevention and treatment of osteoporosis in animal models, there is much diversity among their strains. Since most of these compound strains are naturally present and formulated, there is no consensus on the most effective probiotic compound. Among the studies that examined the effect of probiotic combinations on the prevention and treatment of osteoporosis, we discussed in depth the familiar strains. Despite all the differences and similarities, our research team finally concluded that common strains found in the most effective probiotic combinations for osteoporosis are Lactobacillus acidophilus, Lacticaseibacillus casei, Bifidobacterium longum, Bacillus coagulans, Limosilactobacillus reuteri. Although this conclusion was not based on a systematic review study, it was based on the experiences of expert clinicians in the treatment of osteoporosis and manufacturers of probiotic formulations. In the next step, we divided these strains into several hybrid groups using a mathematical matrix, divided them into seven groups, and examined their preventive effects on osteoporosis in rat. Therefore, we inspected the effectiveness of seven probiotic combinations from five native probiotics (Lactobacillus acidophilus, Limosilactobacillus reuteri, Lacticaseibacillus casei, Bifidobacterium longum, and Bacillus coagulans), as mentioned in Table 1, on BMD, BMC, of global, spine, femur, and tibia on ovariectomized rats.

In this study, combination NO 4, containing L. acidophilus, B. longum, and L. reuteri, is the most influential group on global, spine, and femur BMD. Combination NO 3, containing L. acidophilus, L. casei, and L. reuteri, also significantly affects the BMD of the tibia among the treatment group. According to Kim et al., L. casei considerably improved the tibia BMD in OVX rats [31]. While evaluating the effects of our probiotic combinations on femur, spine, global, and tibia, we indicated that particular combination had the most influence on the special bone type. It was evident that combination NO 4 had the most significant increasing effect on global BMC. Also, combination NO 1 (comprising L. acidophilus, L. casei, and B. longum), NO 6 (containing L. casei, B. longum, and Bacillus coagulans), NO 7 (containing L. casei, L. reuteri, and B. longum), and NO 4 had the most considerable raising effect on spine BMC.

What can be deduced from this study is that L. acidophilus is a common strain in the most effective combination sets tested to improve both BMC and BMC. Our previous study reported that L. acidophilus was more effective in the treatment groups in the case of global, spine, and femur BMD than the OVX untreated group [3]. Some studies reported that L. reuteri might decrease fracture, increase BMD, BMC, and trabecular number and thickness and weaken the trabecular space of the vertebrae and femurs. Inflammation may be the leading cause of abnormal bone regeneration and the onset of bone loss. Some studies have shown that increased inflammatory cytokines are associated with osteoclastic bone resorption, low bone mineral density (BMD), elevated bone resorption, and increased fracture risk. Therefore, blockade of the pro-inflammatory cytokine levels due to intake of probiotic supplements leads to a decrease in bone resorption [32, 33]. We also observed that this strain significantly improved global BMD, BMC, femur BMD, and BMC [3].

Also, B. longum is present in many effective combinations of this study, especially NO 4. Under our previous studies, B. longum could successfully affect femur BMD and BMC. The impact of B. longum on bone density, bone mineral content, bone remodeling, bone structure, and osteoclast/osteoblast gene expression markers was previously evaluated in the OVX rat model for 16 weeks and improved their bone density, trabecular number, thickness, and femoral strength. B. longum likewise reduces levels of serum C-terminal telopeptide [34].

The current study disclosed an apparent synergistic effect between Lactobacillus species and B. longum. Evidence studies in preclinical suggest that dual colonization of Lactobacillus sp. and Bifidobacterium sp. ameliorated severe diarrhea and synergistically decreased virus shedding titers probably because of modulating mucosal and systemic innate and adaptive immunity [35]. This synergistic effect may also be conceivable for the treatment and prevention of osteoporosis. Also, Lactobacillus sp. and Bifidobacterium sp. combination synergistically alleviated immobilization stress and anxiety behaviors, nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) activation, brain-derived neurotrophic factor (BDNF) expression, tumor necrosis factor (TNF)-α, interleukin (IL)- 6, and lipopolysaccharide levels via maintaining of the gut immune responses and microbiota arrangement [36]. Additionally, these strains repressed the expression of NF-κB activation and TNF-α. Some recent studies indicated such an effect on bone mineralization when naturally combined Lactobacillus sp. and Bifidobacterium sp., attributed to changes in gut microbiota and ecology [37]. Most studies that have evaluated the effectiveness of probiotic compounds include Lactobacillus sp. In some studies, however, Bifidobacterium sp. was used in combination with Lactobacillus. This indicates the greater effectiveness of lactobacilli sp. in a mixture. Bifidobacterium sp. was used in most studies that did not show a more significant effect of the probiotic combinations, indicating that the therapeutic effect of Bifidobacterium sp. may be repressed when other species exist in a multivalent probiotic mixture. Among the studies in which the mixture was not more effective, all contained one or more species of lactobacilli along with Bifidobacterium sp. and several other genera. This reduction in effect indicates that sometimes the high diversity of strains in a probiotic combination diminishes the effectiveness of a multivariate probiotic. These diverse strains appear to constrain each other in the body environment, possibly due to the secretion of antagonistic agents or struggle for nutrients or receptors in the gastrointestinal tract. Our previous study revealed that oral administration of B. longum or Lactobacillus strains declines BMD and BMC criteria induced thorough ovariectomizing in response to hormone deficiency. However, the mixture of these probiotics was more effective in osteoporotic rats at all levels of BMD and BMC. This study’s results indicated that probiotic’s ameliorative effect on bone loss in ovariectomized rat model is strain-specific. The consequences have also revealed a discrepancy concerning the influence of these different probiotic treatments on releasing plasma levels of hormones. It seems that the L. acidophilus strain, combined with L. casei and B. longum strains, regulated immune systems, increased calcium absorption, and even ameliorated hormone levels under postmenopausal like osteoporosis conditions. The role of B. longum in the mixture of other strains can be elucidated by its anti-inflammatory features. The action of B. longum depends on the inflammatory state of the intestine, and it induces an anti-inflammatory response in inflammatory bowel disease. The anti-inflammatory properties of B. longum may be due to its capability to prevent the binding of pathogens to epithelial cells. Considering the functional specificity of each strain at the peripheral and central levels, all these data demonstrate that CNS function is improved by a mixture of these two or three probiotics (L. Acidophilus, B. Longum, and L. casei). This combination of probiotics is feasible and acceptable to people worldwide with no severe side effects. Although probiotic nutraceutical products have been extensively used for a long time, the use of probiotics to improve general health is increasing. For example, in a recent data network survey, approximately 40% of people indicated that in 2018 they used probiotics in various forms. Probiotic combinations are easy to process, formulate, commercialize, and prescribe in commercial conditions. Three effective probiotic strains in this study are available in various dairy products and other human fermented foods. They can be easily isolated and purified using conventional microbiological methods. Because of the growing interest in probiotics and their association with osteoporosis and bone loss, it is interesting to analyze the cost-effectiveness ratio of this potential intervention for osteoporosis.

However, the cost-effectiveness ratio of probiotic combinations for osteoporosis has not been estimated in the literature. Therefore, evaluating the cost-effectiveness of using probiotic combinations to prevent osteoporosis in postmenopausal women can lead to valuable results.

The limitation of our study was that we did not detect the changes of related metabolites of intestinal flora, metabolic indexes of bacterial flora and bone related pathway proteins, bone remodeling related indexes, and bone microstructure.

Conclusion

Our observations revealed that probiotic combinations could be used for bone formation improvement, bone resorption reduction, and microstructure of femur changing. Among seven combined effective probiotics, a combination containing L. acidophilus, B. longum, and L. reuteri is the most influential group in ovariectomized osteoporotic rat. This combination synergistically increased the level of BMC and BMD together with calcium absorption, osteoblast activity, and vitamin D concentration that cause elevated bone health. Further clinical studies are needed to ensure that these reports are qualified for human beings.

Availability of data and materials

The datasets generated during and/or analyzed during the current study are available from the corresponding author on reasonable request.

References

  1. Akkawi I, Zmerly H. Osteoporosis: current concepts. Joints. 2018;6(02):122–7. https://doi.org/10.1055/s-0038-1660790.

    Article  PubMed  PubMed Central  Google Scholar 

  2. Montazeri-Najafabady N, Ghasemi Y, Dabbaghmanesh MH, Ashoori Y, Talezadeh P, Koohpeyma F, et al. Exploring the bone sparing effects of postbiotics in the postmenopausal rat model. BMC Complement Med Ther. 2021;21(1):1–12. https://doi.org/10.1186/s12906-021-03327-w.

    Article  CAS  Google Scholar 

  3. Montazeri-Najafabady N, Ghasemi Y, Dabbaghmanesh MH, Talezadeh P, Koohpeyma F, Gholami A. Supportive role of probiotic strains in protecting rats from ovariectomy-induced cortical bone loss. Probiotics Antimicrob Proteins. 2019;11(4):1145–54. https://doi.org/10.1007/s12602-018-9443-6.

    Article  CAS  PubMed  Google Scholar 

  4. Kuo T-R, Chen C-H. Bone biomarker for the clinical assessment of osteoporosis: recent developments and future perspectives. Biomarker Res. 2017;5(1):1–9. https://doi.org/10.1186/s40364-017-0097-4.

    Article  Google Scholar 

  5. Mohkam M, Rasoul-Amini S, Shokri D, Berenjian A, Rahimi F, Sadraeian M, et al. Characterization and in vitro probiotic assessment of potential indigenous Bacillus strains isolated from soil rhizosphere. Minerva Biotecnol. 2016;28(1):19–28.

    Google Scholar 

  6. Ashoori Y, Mohkam M, Heidari R, Abootalebi SN, Mousavi SM, Hashemi SA, et al. Development and in vivo characterization of probiotic lysate-treated chitosan nanogel as a novel biocompatible formulation for wound healing. Biomed Res Int. 2020;2020:8868618. https://doi.org/10.1155/2020/8868618.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Azarang A, Farshad O, Ommati MM, Jamshidzadeh A, Heidari R, Abootalebi SN, et al. Protective role of probiotic supplements in hepatic steatosis: a rat model study. Biomed Res Int. 2020;2020:5487659. https://doi.org/10.1155/2020/5487659.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Golkar N, Ashoori Y, Heidari R, Omidifar N, Abootalebi SN, Mohkam M, et al. A novel effective formulation of bioactive compounds for wound healing: preparation, in vivo characterization, and comparison of various postbiotics cold creams in a rat model. Evid Based Complement Alternat Med. 2021;2021:8577116. https://doi.org/10.1155/2021/8577116.

    Article  PubMed  PubMed Central  Google Scholar 

  9. Hill C, Guarner F, Reid G, Gibson GR, Merenstein DJ, Pot B, et al. Expert consensus document: the International Scientific Association for Probiotics and Prebiotics consensus statement on the scope and appropriate use of the term probiotic. Nat Rev Gastroenterol Hepatol. 2014. https://doi.org/10.1038/nrgastro.2014.66.

    Article  PubMed  Google Scholar 

  10. McCabe L, Britton RA, Parameswaran N. Prebiotic and probiotic regulation of bone health: role of the intestine and its microbiome. Curr Osteoporos Rep. 2015;13(6):363–71. https://doi.org/10.1007/s11914-015-0292-x.

    Article  PubMed  PubMed Central  Google Scholar 

  11. Rodrigues FC, Castro ASB, Rodrigues VC, Fernandes SA, Fontes EAF, de Oliveira TT, et al. Yacon flour and Bifidobacterium longum modulate bone health in rats. J Med Food. 2012;15(7):664–70. https://doi.org/10.1089/jmf.2011.0296.

    Article  CAS  PubMed  Google Scholar 

  12. Yang L-C, Lin S-W, Li I, Chen Y-P, Tzu S-Y, Chou W, et al. Lactobacillus plantarum GKM3 and Lactobacillus paracasei GKS6 supplementation ameliorates bone loss in ovariectomized rat by promoting osteoblast differentiation and inhibiting osteoclast formation. Nutrients. 2020;12(7):1914. https://doi.org/10.3390/nu12071914.

    Article  CAS  PubMed Central  Google Scholar 

  13. Dar HY, Shukla P, Mishra PK, Anupam R, Mondal RK, Tomar GB, et al. Lactobacillus acidophilus inhibits bone loss and increases bone heterogeneity in osteoporotic rat via modulating Treg-Th17 cell balance. Bone Rep. 2018;8:46–56. https://doi.org/10.1016/j.bonr.2018.02.001.

    Article  PubMed  PubMed Central  Google Scholar 

  14. Hemarajata P, Versalovic J. Effects of probiotics on gut microbiota: mechanisms of intestinal immunomodulation and neuromodulation. Therap Adv Gastroenterol. 2013;6(1):39–51. https://doi.org/10.1177/1756283X12459294.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Scholz-Ahrens KE, Ade P, Marten B, Weber P, Timm W, AÏ‚il Y, et al. Prebiotics, probiotics, and synbiotics affect mineral absorption, bone mineral content, and bone structure. J Nutr. 2007;137(3):838S-S846. https://doi.org/10.1093/jn/137.3.838S.

    Article  CAS  PubMed  Google Scholar 

  16. Legette LL, Lee W-H, Martin BR, Story JA, Arabshahi A, Barnes S, et al. Genistein, a phytoestrogen, improves total cholesterol and Synergy®, a prebiotic, improved calcium utilization but there were no synergistic effects. Menopause (New York, NY). 2011;18(8):923. https://doi.org/10.1097/gme.0b013e3182116e81.

    Article  Google Scholar 

  17. Parvaneh K, Jamaluddin R, Karimi G, Erfani R. Effect of probiotics supplementation on bone mineral content and bone mass density. Sci World J. 2014;2014. https://doi.org/10.1155/2014/595962

  18. Chapman CM, Gibson GR, Rowland I. Health benefits of probiotics: are mixtures more effective than single strains? Eur J Nutr. 2011;50(1):1–17. https://doi.org/10.1007/s00394-010-0166-z.

    Article  CAS  PubMed  Google Scholar 

  19. Quach D, Britton RA. Gut microbiota and bone health.  In: McCabe L Parameswaran N. (eds)Understanding the Gut-Bone Signaling Axis. Advances in Experimental Medicine and Biology. Cham: Springer; 2017;1033:47–58.https://doi.org/10.1007/978-3-319-66653-2_4. 

  20. Ding K, Hua F, Ding W. Gut microbiome and osteoporosis. Aging Dis. 2020;11(2):438. https://doi.org/10.14336/AD.2019.0523.

    Article  PubMed  PubMed Central  Google Scholar 

  21. Baggio LL, Drucker DJ. Biology of incretins: GLP-1 and GIP. Gastroenterology. 2007;132(6):2131–57.

    Article  CAS  Google Scholar 

  22. Yadav VK, Ryu J-H, Suda N, Tanaka KF, Gingrich JA, Schütz G, et al. Lrp5 controls bone formation by inhibiting serotonin synthesis in the duodenum. Cell. 2008;135(5):825–37.

    Article  CAS  Google Scholar 

  23. Sjögren K, Engdahl C, Henning P, Lerner UH, Tremaroli V, Lagerquist MK, et al. The gut microbiota regulates bone mass in mice. J Bone Miner Res. 2012;27(6):1357–67.

    Article  Google Scholar 

  24. Ghanem K, Badawy I, Abdel-Salam A. Influence of yoghurt and probiotic yoghurt on the absorption of calcium, magnesium, iron and bone mineralization in rats. Milchwissenschaft. 2004;59(9–10):472–5.

    CAS  Google Scholar 

  25. Kruger MC, Fear A, Chua W-H, Plimmer GG, Schollum LM. The effect of Lactobacillus rhamnosus HN001 on mineral absorption and bone health in growing male and ovariectomised female rats. Dairy science & technology. 2009;89(3):219–31.

    Article  CAS  Google Scholar 

  26. Tomofuji T, Ekuni D, Azuma T, Irie K, Endo Y, Yamamoto T, et al. Supplementation of broccoli or Bifidobacterium longum–fermented broccoli suppresses serum lipid peroxidation and osteoclast differentiation on alveolar bone surface in rats fed a high-cholesterol diet. Nutr Res. 2012;32(4):301–7.

    Article  CAS  Google Scholar 

  27. Ouwehand AC, Invernici MM, Furlaneto FA, Messora MR. Effectiveness of multi-strain versus single-strain probiotics: current status and recommendations for the future. J Clin Gastroenterol. 2018;52:S35–40. https://doi.org/10.1097/MCG.0000000000001052.

    Article  CAS  PubMed  Google Scholar 

  28. Li JY, Chassaing B, Tyagi AM, Vaccaro C, Luo T, Adams J, et al. Sex steroid deficiency-associated bone loss is microbiota dependent and prevented by probiotics. J Clin Invest. 2016;126(6):2049–63. https://doi.org/10.1172/jci86062.

    Article  PubMed  PubMed Central  Google Scholar 

  29. Caballero-Franco C, Keller K, De Simone C, Chadee K. The VSL#3 probiotic formula induces mucin gene expression and secretion in colonic epithelial cells. Am J Physiol Gastrointest Liver Physiol. 2007;292(1):G315–22. https://doi.org/10.1152/ajpgi.00265.2006.

    Article  CAS  PubMed  Google Scholar 

  30. Collins FL, Rios-Arce ND, Schepper JD, Parameswaran N, McCabe LR. The potential of probiotics as a therapy for osteoporosis. Microbiol Spectr. 2017;5(4):10.1128/microbiolspec.BAD-0015–2016. https://doi.org/10.1128/microbiolspec.BAD-0015-2016.

    Article  Google Scholar 

  31. Amdekar S, Kumar A, Sharma P, Singh R, Singh V. Lactobacillus protected bone damage and maintained the antioxidant status of liver and kidney homogenates in female wistar rats. Mol Cell Biochem. 2012;368(1):155–65. https://doi.org/10.1007/s11010-012-1354-3.

    Article  CAS  PubMed  Google Scholar 

  32. McCabe LR, Irwin R, Schaefer L, Britton RA. Probiotic use decreases intestinal inflammation and increases bone density in healthy male but not female rat. J Cell Physiol. 2013;228(8):1793–8. https://doi.org/10.1002/jcp.24340.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Ohlsson C, Engdahl C, FÃ¥k F, Andersson A, Windahl SH, Farman HH, et al. Probiotics protect rat from ovariectomy-induced cortical bone loss. PLoS ONE. 2014;9(3):e92368. https://doi.org/10.1002/jcp.24340.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Parvaneh K, Ebrahimi M, Sabran MR, Karimi G, Hwei AN, Abdul-Majeed S, et al. Probiotics (Bifidobacterium longum) increase bone mass density and upregulate Sparc and Bmp-2 genes in rats with bone loss resulting from ovariectomy. Biomed Res Int. 2015;2015:897639. https://doi.org/10.1155/2015/897639.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Vlasova AN, Kandasamy S, Chattha KS, Rajashekara G, Saif LJ. Comparison of probiotic lactobacilli and bifidobacteria effects, immune responses and rotavirus vaccines and infection in different host species. Vet Immunol Immunopathol. 2016;172:72–84. https://doi.org/10.1016/j.vetimm.2016.01.003.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Jang H-M, Lee K-E, Kim D-H. The preventive and curative effects of Lactobacillus reuteri NK33 and Bifidobacterium adolescentis NK98 on immobilization stress-induced anxiety/depression and colitis in rat. Nutrients. 2019;11(4):819. https://doi.org/10.3390/nu11040819.

    Article  CAS  PubMed Central  Google Scholar 

  37. Scholz-Ahrens KE, Adolphi B, Rochat F, Barclay DV, de Vrese M, Açil Y, et al. Effects of probiotics, prebiotics, and synbiotics on mineral metabolism in ovariectomized rats — impact of bacterial mass, intestinal absorptive area and reduction of bone turnover. NFS J. 2016;3:41–50. https://doi.org/10.1016/j.nfs.2016.03.001.

    Article  Google Scholar 

Download references

Acknowledgements

The authors would like to thank the Vice Chancellery of Shiraz University of Medical Sciences.

Funding

This study was supported by Shiraz University of Medical Sciences by the project number 97–01-33–18906.

Author information

Authors and Affiliations

Authors

Contributions

AG contributed to the design and implementation of the research, to the analysis of the results and to the writing of the manuscript. MHD contributed to the design and implementation of the research, and to the writing of the manuscript. YG contributed to the design and implementation of the research, and to the writing of the manuscript. FK contributed to the design and implementation of the research, performed the experiments, analyzed the results and wrote of the manuscript. PT performed the experiments, analyzed the results and wrote of the manuscript. NMN contributed to the design and implementation of the research, to the analysis of the results and to the writing of the manuscript. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Nima Montazeri-Najafabady.

Ethics declarations

Ethics approval and consent to participate

All experimental protocols were approved by the Ethics Committee (NO. IR.SUMS.REC.1398.500) of Shiraz University of Medical Sciences, Shiraz, Iran.

All methods were carried out in accordance with relevant guidelines and regulations.

All methods are reported in accordance with ARRIVE guidelines (https://arriveguidelines.org) for the reporting of animal experiments.

Consent for publication

Not applicable.

Competing interests

The authors have no competing interests to declare that are relevant to the content of this article.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Gholami, A., Dabbaghmanesh, M.H., Ghasemi, Y. et al. The ameliorative role of specific probiotic combinations on bone loss in the ovariectomized rat model. BMC Complement Med Ther 22, 241 (2022). https://doi.org/10.1186/s12906-022-03713-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12906-022-03713-y

Keywords